HK1075677B - Effectors of innate immunity - Google Patents
Effectors of innate immunity Download PDFInfo
- Publication number
- HK1075677B HK1075677B HK05107785.9A HK05107785A HK1075677B HK 1075677 B HK1075677 B HK 1075677B HK 05107785 A HK05107785 A HK 05107785A HK 1075677 B HK1075677 B HK 1075677B
- Authority
- HK
- Hong Kong
- Prior art keywords
- seq
- peptide
- peptides
- cells
- cationic
- Prior art date
Links
Description
Information on related applications
This application claims priority from U.S. patent application serial No. 60/336,632, filed on 3.12.2001, in accordance with 35 USC 119(e), which is hereby incorporated by reference in its entirety.
Technical Field
[0001] The present invention relates generally to peptides, and more particularly, to peptides that are therapeutically effective, and that are effective for drug development associated with pathologies caused by microbial infections and that effectively modulate innate immunity or anti-inflammatory activity.
Background
[0002] Infectious diseases are a leading cause of death worldwide. According to the 1999 world health organization, over 1.3 million people die each year from infectious diseases. Infectious diseases are the third leading cause of death in north america, with 20% of deaths annually attributed to infectious diseases and increasing at a rate of 50% since 1980. The key to the success of many medical and surgical treatments is also the control of infectious diseases. The discovery and use of antibiotics has become one of the major achievements in modern medicine. Without antibiotics, physicians are unable to perform complex surgery, chemotherapy, and most medical interventions such as catheterization.
[0003] Antibiotics are currently sold worldwide in $ 260 billion. However, abuse and certain improper use of antibiotics has led to the evolution of new strains that are resistant to antibiotics. Antibiotic resistance has become an integral part of the medical field. Some bacteria, such as vancomycin-resistant enterococci, VRE and methicillin-resistant staphylococci, MRSA strains, cannot be combated with antibiotics, and patients infected with these bacteria often die. The discovery of antibiotics has become one of the most difficult areas for new drug development, and many large pharmaceutical companies have curtailed or completely stopped their antibiotic development programs. However, with the dramatic increase in the problem of antibiotic resistance, including the emergence of infectious diseases that cannot be treated, the medical need for new antimicrobial therapies is clearly unmet and agents that can have an impact on innate immunity will become one of such agents.
[0004] The innate immune system is an extremely effective, highly evolved, universal defense system. Elements of innate immunity (elements) are always present at low levels and when stimulated they are activated very rapidly. Stimulation includes interactions between bacterial signaling molecules and pattern recognition receptors on the surface of body cells, or other pathogenic mechanisms. Every day, humans are exposed to thousands of potentially pathogenic microorganisms through the food and moisture ingested by individuals, the air breathed by individuals, the surfaces touched by individuals, pets and humans. Innate immunity acts to prevent the development of diseases caused by these pathogens. The innate immune system differs from so-called adaptive immunity (which includes antibody and antigen-specific B and T lymphocytes) in that the former is always present, produces an immediate effect, and is relatively non-specific for any particular pathogen. Adaptive immunity requires the amplification of specific recognition elements (recognitions) and therefore takes days to weeks to respond. Even if vaccination prestimulates adaptive immunity, it may take three days or more to respond to a pathogen, however, innate immunity may respond immediately or very quickly (hours). Innate immunity is involved in a variety of effector functions, including phagocytes, complement, and the like, but is not fully understood. In general, many innate immune responses are "triggered" by the binding of microbial signaling molecules to pattern recognition receptors on the surface of host cells, referred to as Toll-like receptors. In an inflammatory response, there are many such effector functions (effector functions) grouped together. However, a too acute inflammatory response can lead to a deleterious response to the body, in extreme cases sepsis can occur, and death is likely.
[0005] During the course of infection, structural components are released from infectious agents (infectious agents), resulting in an inflammatory response that, when uninhibited, may lead to a potentially lethal condition, sepsis. There are approximately 780,000 sepsis patients in north america each year. Sepsis can occur as a result of infectious diseases acquired in the community, such as pneumonia, and can be a complication in the treatment of trauma, cancer, or major surgery. When the body is completely unable to control the inflammatory response, severe sepsis occurs and the body organs begin to fail. As many as 120,000 people die of sepsis annually in north america. Sepsis may also involve pathogenic microorganisms or toxins in the blood (e.g., sepsis), which is a major cause of human death. Gram-negative bacteria are the most prevalent organisms associated with such diseases. However, gram-positive bacteria are also increasingly causing infectious diseases. Both gram negative and gram positive bacteria and their components can cause sepsis.
[0006] The presence of microbial components causes the release of pro-inflammatory cytokines, of which tumor necrosis factor-alpha (TNF-alpha) is of paramount importance. TNF-alpha and other proinflammatory cytokines can in turn lead to the release of other proinflammatory mediators (pro-inflammatory mediators) and to an inflammatory cascade. Gram-negative sepsis is typically caused by the release of lipopolysaccharide (LPS, also known as endotoxin), a component of the bacterial outer membrane. Endotoxins in the blood, known as endotoxemia (endotoxemia), which result primarily from bacterial infections, can be released during antibiotic therapy. Gram-positive sepsis can be caused by the release of bacterial cell wall components such as lipoteichoic acid (LTA), Peptidoglycan (PG), rhamnose-glucose polymers produced by streptococci, or capsular polysaccharides produced by staphylococci. Unlike mammalian DNA, bacterial or other non-mammalian DNA contains a high frequency of unmethylated cytosine-guanosine dimers (CpGDNA), and has been shown to trigger septic conditions as well, including TNF- α production. Mammalian DNA contains CpG dinucleotides much less frequently, and they are often in methylated form. In addition to their natural release during bacterial infection, antibiotic treatment can also result in the release of the bacterial cell wall components LPS and LTA, and possibly also bacterial DNA. This may hinder recovery from infection, or even lead to sepsis.
[0007] Cationic peptides are continuously recognized as a form of defense against infection, although their primary effect is thought to be antimicrobial in the scientific and patent literature (Hancock, R.E.W., and R.Lehrer.1998 Cationic peptides: and inorganic sources of infection. trends in Biotechnology 16: 82-88). Cationic peptides with antimicrobial activity have been isolated from a variety of organisms. In fact, these peptides provide a mechanism for defense against microorganisms such as bacteria and yeast. Generally, these cationic peptides are thought to interact with the cytoplasmic membrane and, in most cases, form channels and lesions, thereby exerting antimicrobial activity against bacteria. In gram-negative bacteria, they act with LPS to make the outer membrane permeable, thereby leading to self-promoted uptake across the outer membrane and to the cytoplasmic membrane. Examples of cationic antimicrobial peptides include indolicidin (antimicrobial polypeptide from bovine neutrophils), defensins (defensins), cecropins (cecropins) and magainins.
[0008]Recently, it has become increasingly recognized that in other aspects of innate immunity, these peptides can act as effectors (Hancock, r.e.w. and g.diamond.2000.the role of cationic peptides in amino acid host reactions. trends in Microbiology)8:402-410.;Hancock,R.E.W.2001.Cationic peptides:effectors in innate immunity andnovel antimicrobials.Lancet Infectious Diseases 1: 156-164), although it is not known whether the antimicrobial function and effector function are related.
[0009] Some cationic peptides have an adsorption effect that can bind bacterial products such as LPS and LTA. These cationic peptides are capable of inhibiting the production of cytokines in response to LPS and thus are capable of preventing to some extent lethal shock. However, it has not been demonstrated whether these effects are due to the binding of the peptide to LPS and LTA or to the direct action of the peptide and the host cell. In response to attack by a microorganism or a microbial signalling molecule such as LPS, the cationic peptide is induced by a regulatory pathway similar to that used by the mammalian immune system (involving Toll-like receptors and transcription factors; NF. kappa.B). It appears that cationic peptides have a key role in innate immunity. Mutations affecting the induction of antibacterial peptides can reduce survivors in response to bacterial challenge. Similarly, the sensitivity to lethal fungal infections is increased when the Toll pathway of Drosophila is mutated to reduce expression of antifungal peptides. In humans, patients with granulosa deficiency syndrome are completely deficient in alpha-defensin and suffer from frequent and severe bacterial infections. Other evidence includes that some peptides can be induced by infectious agents (infectious agents), and that the concentration of these peptides has been found to be quite high where inflammation occurs. Cationic peptides may also regulate cell migration to promote the ability of leukocytes to resist bacterial infection. For example, two human α -defensin polypeptides, HNP-1 and HNP-2, have been found to be chemotactic for murine and human T cells and monocytes, and human β -defensins appear to be able to act as chemoattractants (chemoattractants) for immature dendritic cells and memory T cells by acting with CCR 6. Similarly, the porcine cationic peptide PR-39 was found to be chemotactic for neutrophils. However, it is still unclear whether peptides with different structures and compositions share these properties.
[00010] LL-37 is the only known cathelicidin from human (a precursor of antimicrobial peptides with potent membrane activity) produced by bone marrow precursors, testis, human keratinocytes during inflammatory disease and airway epithelial cells. Cathelicidin peptides are characterized by a relatively high proportion of sequence identity in the N-terminal precursor region (prepro region) known as the cathelin domain. Cathelicidin peptides are stored as unactivated pro-peptide precursors and upon stimulation are processed into active peptides.
Summary of The Invention
[00011] The invention is based on the following pioneering findings: novel compounds that block or reduce sepsis and/or inflammation in a subject can be selected based on the pattern of polynucleotide expression that is modulated by endotoxically toxic lipopolysaccharides, lipoteichoic acids, CpG DNA, or other cellular components (e.g., microorganisms or their cellular components), but is affected by the cationic peptide. Furthermore, based on the use of cationic peptides as a tool, selective enhancers of innate immunity can be identified that do not elicit a septic response, but block/suppress the inflammatory and/or septic response.
[00012] Thus, in one embodiment, a method is provided for identifying polynucleotides or patterns of polynucleotides that are modulated by one or more inducers of sepsis or inflammation and that are inhibited by a cationic peptide. The methods of the invention comprise contacting one or more polynucleotides with one or more inducers of sepsis or inflammation and simultaneously or immediately thereafter contacting the one or more polynucleotides with a cationic peptide. Detection of differences in expression, either up-regulation or down-regulation, between the presence and absence of the cationic peptide, changes in expression, can be used as an indication of a polynucleotide or pattern of polynucleotides that is regulated by sepsis or an inducer of inflammation and that is simultaneously inhibited by the cationic peptide. In another aspect, the invention provides one or more polynucleotides identified by the above methods. The sepsis or inflammation modulator comprises LPS, LTA or CpG DNA or microbial components (or any combination thereof), or a related agent.
[0010] In another embodiment, the invention provides a method of identifying an agent that blocks sepsis or inflammation, comprising combining a polynucleotide identified by the foregoing methods with an agent, wherein the expression of the polynucleotide is modulated in the presence of the agent compared to the absence of the agent, and wherein the modulation in expression affects inflammation or a sepsis response.
[0011] In another embodiment, the invention provides a method for identifying a pattern of polynucleotide expression when an inflammatory or septic response is inhibited by 1) contacting cells with LPS, LTA and/or CpG DNA in the presence or absence of a cationic peptide, and 2) detecting the pattern of polynucleotide expression in the presence or absence of the peptide. The pattern obtained in the presence of the peptide represents an inhibition of the inflammatory or septic response. In another aspect, the pattern obtained in the presence of the peptide is compared to the pattern obtained in the presence of a test compound to identify compounds that provide a similar pattern. In another aspect, the invention provides compounds identified by the foregoing methods.
[0012] In another embodiment, the invention provides a method of identifying an agent that potentiates innate immunity comprising contacting one or more polynucleotides encoding polypeptides involved in innate immunity with an agent of interest, wherein expression of the polynucleotide is modulated in the presence of the agent, and the modulated expression results in potentiation of innate immunity, as compared to the absence of the agent. Preferably, the agent does not stimulate a septic response in the individual. In one aspect, the agent increases the expression of an anti-inflammatory polynucleotide. Exemplary, but non-limiting, anti-inflammatory polynucleotides encode proteins such as: IL-1R antagonist homolog 1(AI167887), IL-10 Rbeta (AA486393), IL-10 Ralpha (U00672), TNF receptor member 1B (AA150416), TNF receptor member 5(H98636), TNF receptor member 11B (AA194983), HLA II IK cytokine down-regulator (R39227), TGF-B inducible early growth response protein 2(AI473938), CD2(AA927710), IL-19 (NM-013371), or IL-10 (M57627). In one aspect, the agent reduces expression of a polynucleotide encoding a proteasome subunit involved in NK-kb activation, e.g., proteasome subunit 26S (NM — 013371). In one aspect, the agent can act as an antagonist of a protein kinase. In one aspect, the agent is a nucleic acid sequence selected from SEQ ID NOs: 4-54.
[0013] In another embodiment, the invention provides a method for identifying a pattern of polynucleotide expression to identify compounds that selectively potentiate innate immunity. The invention includes detecting a pattern of polynucleotide expression in cells contacted with and not contacted with a cationic peptide, wherein the pattern in the presence of the peptide represents stimulation of innate immunity; detecting a pattern of polynucleotide expression from the cells contacted with a test compound, wherein the test compound enhances innate immunity if the pattern is similar to that observed in the presence of the cationic peptide when the test compound is used. Preferably, the compound does not stimulate a septic response in the individual.
[0014] In another embodiment, the invention provides a method of inferring the infection status of a mammalian subject from a nucleic acid sample from the subject by determining the expression pattern of polynucleotides in the nucleic acid sample, e.g., an increased expression of at least two polynucleotides in tables 50, 51, and/or 52 as compared to an uninfected subject. Also included are polynucleotide expression profiles obtained by any of the above methods.
[00013] In another aspect, cationic peptides are provided that are antagonists of CXCR-4. In yet another aspect, there is provided a method of identifying a cationic peptide that is an antagonist of CXCR-4, comprising contacting a T cell with SDF-1 in the presence or absence of a test peptide, and measuring chemotaxis. A test peptide, if it has reduced chemotaxis in the presence of the peptide, indicates that the peptide is an antagonist of CXCR-4. The cationic peptide may also function to reduce expression of the SDF-1 receptor polynucleotide (NM-013371).
[0015] In all of the above methods, the compounds or agents of the invention include, but are not limited to, peptides (peptides), cationic peptides (cationic peptides), peptidomimetics (peptidomimetics), chemical compounds (chemical compounds), polypeptides (polypetides), nucleic acid molecules (nucleic acids molecules), and the like.
[0016] In yet another aspect, the present invention provides isolated cationic peptides. The isolated cationic peptides of the invention can be represented by any of the following general formulas and one letter amino acid notation:
X1X2X3IX4PX4IPX5X2X1(SEQ ID NO: 4) wherein X1Is one or two of R, L or K, X2Is C, S or A, X3Is an R or P, X4Is an A or V, X5Is a V or W;
X1LX2X3KX4X2X5X3PX3X1(SEQ ID NO: 11), wherein X1Is one or two of D, E, S, T or N, X2Is one or two of P, G or D, X3Is an G, A, V, L, I or Y, X4Is an R, K or H, X5Is an S, T, C, M or R;
X1X2X3X4WX4WX4X5k (SEQ ID NO: 18), wherein X1Is one to four amino acids selected from A, P or R, X2Is one or two aromatic amino acids (F, Y and W), X3Is a P or K, X4Is zero to two amino acids selected from A, P, Y or W, X5Is one to three amino acids selected from R or P;
X1X2X3X4X1VX3X4RGX4X3X4X1X3X1(SEQ ID NO: 25), wherein X1Is one or two of R or K, X2Is a polar or charged amino acid (S, T, M, N, Q, D, E, K, R and H), X3Is C, S, M, D or A, X4Is F, I, V, M or R;
X1X2X3X4X1VX5X4RGX4X5X4X1X3X1(SEQ ID NO: 32) wherein X1Is one or two of R or K, X2Is a polar or charged amino acid (S, T, M, N, Q, D, E, K, R andH),X3is C, S, M, D or A, X4Is an F, I, V, M or R, X5Is an A, I, S, M, D or R; and
KX1KX2FX2KMLMX2ALKKX3(SEQ ID NO: 39) wherein X1Is a polar amino acid (C, S, T, M, N and Q); x2Is A, L, S or K, X3Is 1 to 17 amino acids selected from G, A, V, L, I, P, F, S, T, K and H;
KWKX2X1X1X2X2X1X2X2X1X1X2X2IFHTALKPISS (SEQ ID NO: 46), wherein X1Is a hydrophobic amino acid, X2Is a hydrophilic amino acid.
[0017] In addition, in another aspect the invention provides isolated cationic peptides KWKSFLRTFKSPVRTVFHTALKPISS (SEQ ID NO: 53) and KWKSYAHTIMSPVRLVFHTALKPISS (SEQ ID NO: 54).
[0018] Also provided are nucleic acid sequences encoding the cationic peptides of the invention, vectors comprising these polynucleotides, and host cells containing these vectors.
Detailed Description
[0019] The present invention provides novel cationic polypeptides that can be characterized by a set of general formulae that are capable of modulating (e.g., up-regulating and/or down-regulating) expression of a polynucleotide, thereby modulating and/or modulating an inflammatory response and/or innate immunity.
[0020] As used herein, "innate immunity" refers to a biological defense against pathogen invasion, protecting the innate ability of the organism. Pathogens or microorganisms as used herein may include, but are not limited to, bacteria, fungi, parasites, and viruses. Innate immunity differs from adaptive/adaptive immunity, in which organisms develop their own defense mechanisms, primarily based on antibodies and/or immune lymphocytes, characterized by specificity, scalability and recognition of self versus non-self. Innate immunity provides extensive nonspecific immunity and has no immunological memory for previous exposure. Innate immunity is characterized by the ability to effectively combat a wide range of potential pathogens, independent of prior exposure to a pathogen, and innate immunity is further characterized by the ability to take effect rapidly (the elicitation of a specific immune response takes days to weeks as compared to a specific immune response). Moreover, innate immunity includes immune responses that affect other diseases such as cancer, inflammatory diseases, multiple sclerosis, various viral infections, and the like.
[0021] As used herein, the term "cationic peptide" refers to an amino acid sequence of about 5 to about 50 amino acids in length. In one aspect, the cationic peptides of the invention are from about 10 to about 35 amino acids in length. A peptide is considered a "cationic peptide" if it has enough positively charged amino acids that the pI is greater than 9.0, where pI (isoelectric point) ═ the pH value when the net charge of the peptide is neutral. Typically, at least two of the amino acid residues of the cationic peptide are positively charged, such as lysine and arginine. "positively charged" refers to an amino acid residue side chain having a net positive charge at pH 7.0. Examples of naturally occurring cationic antimicrobial peptides include defensins, cathelicidins, magainins, melittin, cecropin, bactenecins (antimicrobial polypeptides from bovine neutrophils), indolicidins, polyphemusins, tacyphesins (antimicrobial polypeptides from Calla cheilosa blood cells), and analogs thereof, which can be recombinantly produced according to the invention. Many organisms produce cationic peptides and use these molecules as part of a non-specific defense mechanism against microorganisms. These isolated peptides are toxic to a wide variety of microorganisms, including bacteria, fungi, and certain enveloped viruses. When cationic peptides exert a resistance against many pathogens, significant abnormalities and varying degrees of toxicity are present. However, this patent discloses other cationic peptides that are not toxic to microorganisms but are capable of infecting by stimulating innate immune defenses, and the invention is not limited to cationic peptides having antimicrobial activity. In fact, many of the peptides used in the present invention do not have antimicrobial activity.
[0022] Cationic peptides known in the art include, for example, human cathelicidin il-37, the bovine neutrophil polypeptide indolicidin, and variants of bovine bactenecin, Bac 2A.
IL-37 LLGDFFRKSKEKIGKEFKRIVQRIKDFLRNLVPRTES(SEQ ID:1)
Indolicidin ILPWKWPWWPWRR-NH2(SEQ ID NO:2)
Bac2A RLARIVVIRVAR-NH2(SEQ ID NO:3)
[0023] In innate immunity, the immune response is not antigen-dependent. The innate immune process may include the production of secreted molecules and cellular components as previously set forth. In innate immunity, pathogens are recognized by receptors encoded in the germ line. These Toll-like receptors have a wide range of specificities and are capable of recognizing many pathogens. When cationic peptides are present in the immune response, they assist the host in responding to pathogens. This change in the immune response induces the release of chemokines that cause immune cells to concentrate to the site where the infection occurs.
[0024] Chemokines or chemically induced cytokines belong to a class of immune factors that mediate chemotaxis and other proinflammatory phenomena (see, Schall, 1991, Cytokine 3: 165-183). Chemokines are small molecules about 70-80 residues in length, generally divided into two subclasses, an alpha subclass with an N-terminal cysteine (C × C) separated by a single amino acid, and a beta subclass with two adjacent cysteines (CC) at the N-terminus. RANTES, MIP-1 α and MIP-1 β belong to the β subclass (see for review: Horuk, R., 1994, Trends Pharmacol. Sci, 15: 159-. The amino-terminal ends of the beta chemokines RANTES, MCP-1 and MCP-3 are involved in mediating cell migration and the modulation of inflammatory responses induced by these chemokines. This association is observed experimentally that elimination of 8 residues at the amino terminus of MCP-1, 9 residues at the amino terminus of MCP-3 and 8 residues at the amino terminus of RANTES, and addition of a methionine to the amino terminus of RANTES, antagonizes chemotaxis, calcium transfer and/or enzyme release stimulated by their natural counterparts (Gong et al, 1996 J.biol.chem.271: 10521-. In addition, chemotactic activity similar to that of the a-type chemokine can be introduced into MCP-1 by double mutating Tyr 28 and Arg 30 to leucine and valine, suggesting that the internal region of the protein also plays a role in regulating chemotactic activity (Beall et al, 1992, J.biol.chem.267: 3455-) -3459).
[0025] All monomeric forms of chemokines characterized to date have significant structural homology, despite differences in the quaternary structure of the α and β forms. The monomeric structures of the beta and alpha chemokines are very similar, but the dimeric structures of these two types are completely different. Another chemokine, lymphotactin (lymphotactin), which has only one N-terminal cysteine, has also been identified and represents a third subfamily of chemokines (. gamma.) (Yoshida et al, 1995, FEBS Lett.360: 155-.
[0026] Receptors for chemokines belong to the large family of receptors (GCR's) with 7 transmembrane domains coupled to G proteins (see review: Horuk, R., 1994, Trends Pharmacol. Sci.15: 159-165; and Murphy, P. M., 1994, Annu. Rev. immunol. 12: 593-633). Competitive binding and cross-desensitization studies have shown that chemokine receptors have significant promiscuity in binding ligands. Examples of evidence of heterozygosity among beta chemokines include: CC CKR-1 binds RANTES and MIP-1 α (Neote et al, 1993, Cell 72: 415-. Erythrocytes have a receptor (known as Duffy antigen) capable of binding alpha and beta chemokines (Horuk et al, 1994, J.biol.chem.269: 17730-. Thus, significant sequence and structural homology between chemokines and their receptors allows for overlap in receptor-ligand interactions.
[0027] In one aspect, the invention provides the use of compounds comprising the cationic peptides of the invention to reduce sepsis and inflammatory responses by acting directly on host cells. In this aspect, a method of identifying a polynucleotide that is modulated by one or more inducers of sepsis or inflammation is provided, wherein the modulation is altered by a cationic peptide. The sepsis or inflammation inducers include, but are not limited to, endotoxic Lipopolysaccharide (LPS), lipoteichoic acid (LTA) and/or CpG DNA or intact bacteria or other bacterial components. The method of identification relies on contacting the polynucleotide with an inducer of sepsis or inflammation and simultaneously or immediately thereafter with a cationic peptide. An observation of the expression of the polynucleotide in the presence and absence of the cationic peptide, a change in expression indicating that the nucleotide or nucleotide of that type is regulated by an inducer of sepsis or inflammation and inhibited by the cationic peptide. In another aspect, the invention provides polynucleotides identified by the method.
[0028] Once identified, such polynucleotides can be used to screen for compounds that can prevent sepsis or inflammation by affecting the expression of the polynucleotide. Such effects on expression may be up regulation or down regulation of expression. The invention also provides a method of identifying innate immunity enhancers by identifying compounds that do not provoke a septic response and compounds that are capable of preventing or inhibiting an inflammatory or septic response. In addition, the present invention provides compounds used and identified in the above methods.
[0029] Candidate compounds are obtained from a wide variety of sources, including various synthetic and natural compounds. For example, a number of methods can be used to randomly and directionally synthesize various organic compounds and biomolecules, including expression of random oligonucleotides and oligomeric polypeptides. Alternatively, libraries of natural compounds (libraries) in the form of bacterial, fungal, plant and animal extracts may be utilised or conveniently produced. In addition, natural or synthetically produced libraries and compounds may be readily modified by conventional chemical, physical and biochemical means, and combinatorial libraries may also be generated using these libraries and compounds. Known pharmacological agents may be subjected to directed or random chemical modifications, such as acylation, alkylation, esterification, amidation, and the like, to yield structural analogs. Candidate agents may also be found from biomolecules, including but not limited to: peptides, peptidomimetics, carbohydrates, fatty acids, steroids, purines, pyrimidines, polypeptides, polynucleotides, chemical compounds, derivatives (derivatives), structural analogs, or combinations thereof (combinations).
[0030] The incubation elements (components) of the screening assay include conditions that allow the test compound and the polynucleotide of interest to be contacted with each other. The contacting can be in a liquid phase, a solid phase, or in a cell. To screen multiple compounds, the test compounds can be selected as a combinatorial library. Compounds identified according to the methods of the invention can be further evaluated, detected, cloned, sequenced, etc., in solution or after binding to a solid support, by any method commonly used in detecting compounds.
[0031] Generally, in the methods of the invention, cationic peptides are used to detect and localize polynucleotides necessary in the course of sepsis and inflammation. Once identified, the expression pattern (pattern) of the polynucleotide can be obtained by observing its expression in the presence and absence of the cationic peptide. The pattern obtained in the presence of the cationic peptide is also useful for identifying other compounds that inhibit the expression of the polynucleotide and thereby prevent sepsis or inflammation. It is well known to those skilled in the art that non-peptide compounds and peptidomimetics are capable of mimicking the ability of peptides to bind to receptors and enzyme binding sites, and thus can be used to block or elicit biological responses. Where an additional compound of interest provides a similar pattern of polynucleotide expression as in the presence of the cationic peptide, the compound may also be used to modulate sepsis or innate immune response. In this way, the cationic peptides of the invention, while acting as known inhibitors of sepsis and inflammation and known boosters of innate immunity, can also be used as tools to identify other compounds that inhibit sepsis and inflammation and boost innate immunity.
[0032] As will be seen in the examples which follow, the peptides of the invention have the ability to broadly reduce the expression of polynucleotides regulated by LPS. Many of the symptoms seen during severe infection or inflammation, such as fever and elevated white blood cell numbers, are due to high levels of endotoxins in the blood. Endotoxin is a component of the cell wall of gram-negative bacteria and is a pathophysiologically potent initiator of sepsis (trigger). The basic mechanisms of inflammation and sepsis are linked. In example 1, a polynucleotide array was used to determine the effect of cationic peptides on the transcriptional response of epithelial cells. In particular, effects on more than 14,000 different specific polynucleotides induced by LPS were observed. The table shows the changes seen for the peptide-treated cells compared to the control cells. The data obtained indicate that the peptide has the ability to reduce the expression of the polynucleotide induced by LPS.
[0033] Similarly, example 2 demonstrates that the peptides of the invention are capable of neutralizing the stimulation of immune cells by gram-positive and gram-negative bacterial products. In addition, it is noted that certain pro-inflammatory polynucleotides are down-regulated by cationic peptides, as set forth in Table 24, such as TLR1(AI339155), TLR2(T57791), TLR5(N41021), TNF receptor associated factor 2(T55353), TNF receptor associated factor 3(AA504259), TNF receptor superfamily member 12(W71984), TNF receptor superfamily member 17(AA987627), small molecule inducible cytokine subfamily B member 6(AI889554), IL-12R beta 2(AA977194), IL-18 receptor 1(AA 244889), and polynucleotides of the inflammatory disorders are up-regulated by cationic polypeptides, as set forth in Table 25, such as IL-1R antagonist homolog 1(AI167887), IL-10R beta (AA486393), TNF receptor member 1B (AA150416), TNF receptor member 5 (H9811B (19411 II), HLA cytokine downregulator (HLA 39227) 227, Early growth response 2(AI473938) or CD2(AA927710) that can be induced by TGF-B. The applicability and use of these results was demonstrated by in vivo use in mice. Example 3 demonstrates that these peptides are substantially non-toxic to the host cell with which they are contacted.
[0034] In example 4, it can be seen that the cationic peptides of the invention can alter polynucleotide expression in macrophages and epithelial cells. The results of this example demonstrate that pro-inflammatory polynucleotides are down-regulated by cationic peptides (Table 24), while anti-inflammatory polynucleotides are up-regulated by cationic peptides (Table 25).
[0035] In another aspect, the invention provides methods of identifying agents that potentiate innate immunity. In this method, a polynucleotide encoding a polypeptide involved in innate immunity in a host cell is contacted with an agent of interest. Determining the expression of the polynucleotide in the presence and absence of the agent. By comparison of expression, specific modulation of expression indicates that innate immunity is enhanced. On the other hand, the agent does not provoke a septic response, as revealed by the lack of up-regulation (upregulation) of the proinflammatory cytokine TNF- α. In yet another aspect, the agent reduces or blocks an inflammatory or septic response. In yet another aspect, the agent reduces the expression of TNF- α and/or interleukins including, but not limited to, IL-1 β, IL-6, IL-12 p40, IL-12 p70, and IL-8.
[0036] In another aspect, the invention provides methods for direct polynucleotide modulation using cationic peptides and the use of compounds, including cationic peptides, in elements (elements) that stimulate innate immunity. In this aspect, the invention provides methods for determining the expression pattern of polynucleotides in order to identify compounds that potentiate innate immunity. In the method of the invention, cells contacted with the cationic peptide and not contacted with the cationic peptide are subjected to an initial detection of the polynucleotide expression pattern. The pattern of polynucleotide expression in the presence of the peptide indicates that innate immunity is stimulated. The expression pattern of the polynucleotide in the presence of the test compound is then determined, wherein the test compound is used to obtain an expression pattern similar to that obtained in the presence of the cationic peptide, indicating that this is a compound that potentiates innate immunity. In another aspect, the invention provides compounds identified in the above methods. In another aspect, the compounds of the invention stimulate the expression of chemokines or chemokine receptors. Chemokines or chemokine receptors may include, but are not limited to CXCR4, CXCR1, CXCR2, CCR2, CCR4, CCR5, CCR6, MIP-1 α, MDC, MIP-3 α, MCP-1, MCP-2, MCP-3, MCP-4, MCP-5, and RANTES. In yet another aspect, the compound is a peptide, a peptidomimetic (peptidomimetic), a chemical compound, or a nucleic acid molecule.
[0037] In yet another aspect, the polynucleotide expression profile comprises expression of a pro-inflammatory polynucleotide. These pro-inflammatory polynucleotides include, but are not limited to, cyclic finger protein 10(D87451), serine/threonine protein kinase MASK (AB040057), KIAA0912 protein (AB020719), KIAA0239 protein (D87076), RAP1, gtpase activator protein 1(M64788), FEM-1-like death receptor binding protein (AB007856), cathepsin S (M90696), hypothetical protein FLJ20308(AK000315), pim-1 oncogene (M54915), proteasome subunit beta-type 5(D29011), KIAA0239 protein (D87076), bronchobronchial mucin 5 subtype B (AJ001403), cAMP response element binding protein bpcrea, integrin alpham (J031925), Rho-associated kinase 2(NM _004850), PTD017 protein (AL050361), unknown genes (AK 03143, AK034348, 1610450, 161991, 031925), and any combination thereof. In yet another aspect, the polynucleotide expression profile includes expression of cell surface receptors including, but not limited to, retinoic acid receptor (X06614), G protein-coupled receptor (Z94155, X81892, U52219, U22491, AF015257, U66579), chemokine (C-C motif) receptor 7(L31584), tumor necrosis factor receptor superfamily member 17(Z29575), interferon gamma receptor 2(U05875), cytokine receptor-like factor 1(AF059293), class I cytokine receptor (AF053004), lectin II (thrombin) receptor-like 2(U92971), leukemia inhibitory factor receptor (NM — 002310), interferon gamma receptor 1(AL 050337).
[0038] As will be illustrated later, e.g., in tables 1-15, the cationic peptides are capable of neutralizing the host response to a signal molecule of an infectious agent and also of altering the transcriptional response of the host cells, primarily by down-regulating the pro-inflammatory response and/or up-regulating the anti-inflammatory response. Example 5 demonstrates that the cationic peptide can assist the host in responding to pathogens by inducing the release of chemokines that promote immune cell aggregation to the site of infection. These results were confirmed by in vivo application in mice.
[0039] As can be seen from the examples that follow, the cationic peptide significantly affects the host response to pathogens in that the cationic peptide induces a selective pro-inflammatory response, such as a response that promotes the aggregation of immune cells to the site of infection, but does not induce significantly harmful pro-inflammatory cytokines, thereby assisting in the regulation of the host immune response. Sepsis appears to be caused in part by an excessive pro-inflammatory response to an infectious agent. Cationic peptides induce anti-inflammatory responses and inhibit certain potentially harmful pro-inflammatory responses, thereby helping the host produce a "balanced" response to pathogens.
[0040] In example 7, selected MAP kinases were analyzed for activation in order to investigate the basic mechanism by which cationic peptides interact with cells to produce these effects. Macrophages activate MEK/ERK kinase in response to bacterial infection. MEK is a MAP kinase which, when activated, phosphorylates the downstream kinase ERK (extracellular regulatory kinase), which then forms dimers and transfers to the nucleus where it activates transcription factors such as ElK-1, thereby altering expression of the polynucleotide. MEK/ERK kinase has been shown to impair replication of Salmonella macrophage (salmonella). Signal transduction (signal transduction) mediated by MEK kinase and NADPH oxidase plays an important role in the innate defense against intracellular pathogens. As shown below, cationic peptides have an effect on bacterial infection by affecting MAP kinases. These cationic peptides can directly affect kinases. Table 21 shows the changes in MAP kinase polynucleotide expression in response to the peptides, but is not limited to these. These kinases include MAP kinase 6(H070920), MAP kinase 5 (W698649), MAP kinase 7(H39192), MAP kinase 12(AI936909), and protein kinase 3 activated by MAP kinase (W68281).
[0041] In another method, the methods of the invention can be used in combination to identify agents with multiple characteristics, i.e., peptides that have anti-inflammatory/anti-sepsis activity and are capable of boosting innate immunity in part by inducing chemokines in vivo.
[0042] In another aspect, the invention provides methods of inferring the infection status of a mammalian subject from a nucleic acid sample from the subject, the methods relying on determining a pattern of polynucleotide expression in the nucleic acid sample, exemplified by increased polynucleotide expression of at least two polynucleotides in Table 55 as compared to an uninfected subject. In another aspect, the invention provides methods of inferring the infection status of a mammalian subject from a nucleic acid sample from the subject, the methods relying on determining a pattern of polynucleotide expression in the nucleic acid sample, exemplified by the expression of polynucleotides in at least two of the polynucleotides in Table 56 or Table 57, as compared to an uninfected subject. In one aspect of the invention, the infectious state is due to infectious agents or signal molecules derived therefrom, such as, but not limited to, gram-negative and gram-positive bacteria, viruses, fungi or parasites. In another aspect, the invention provides a polynucleotide expression profile of an infected individual determined according to the above method. Once determined, such polynucleotides will be useful in diagnosing conditions associated with the presence or activity of these infectious agents or signaling molecules.
[0043] Example 10 illustrates this aspect of the invention. In particular, table 61 demonstrates that both MEK and NADPH oxidase inhibitors limit bacterial replication (infection of macrophages by salmonella typhimurium (s. typhimurium) activates MEK kinase). This is an example of how bacterial survival can be affected by altering host cell signaling molecules.
[0044] In yet another aspect of the invention, compounds are presented that inhibit T cell chemotaxis induced by stromal cell derived factor 1 (SDF-1). Compounds that reduce the expression of SDF-1 receptors have also been proposed. These compounds may also be used as antagonists or inhibitors of CXCR-4. In one aspect, the invention provides cationic peptides that are CXCR-4 antagonists. In another aspect, the invention provides methods for identifying cationic peptides as CXCR-4 antagonists. The method comprises contacting a T cell with SDF-1 in the presence and absence of a test peptide and measuring chemotaxis. In the presence of the test peptide, a decrease in chemotaxis indicates that the peptide is an antagonist of CXCR-4. These compounds and methods are useful in the therapeutic application of HIV patients. These types of compounds and their utility have been demonstrated, for example, in example 11 (see also tables 62, 63). In this example, cationic peptides have been shown to inhibit cell migration (cell migration) as well as antiviral activity.
[0045] In one embodiment, the present invention provides an isolated cationic peptide having an amino acid sequence of the following general formula (general formula a):
X1X2X3IX4PX4IPX5X2X1(SEQ ID NO: 4) wherein X1Is one or two of R, L or K, X2Is C, S or A, X3Is an R or P, X4Is an A or V, X5Is a V or W.
Examples of peptides of the invention include, but are not limited to:
LLCRIVPVIPWCK (SEQ ID NO: 5), LRCPIAPVIPVCKK (SEQ ID NO: 6), KSRIVPAIPVSLL (SEQ ID NO: 7), KKSPIAPAIPWSR (SEQ ID NO: 8), RRARIVPAIPVARR (SEQ ID NO: 9) and LSRIAPAIPWAKL (SEQ ID NO: 10).
[0046] In another embodiment, the present invention provides an isolated linear cationic peptide having an amino acid sequence of the following general formula (general formula B):
X1LX2X3KX4X2X5X3PX3X1(SEQ ID NO: 11), wherein X1Is one or two of D, E, S, T or N, X2Is one or two of P, G or D, X3Is an G, A, V, L, I or Y, X4Is an R, K or H, X5Is an S, T, C, M or R. Examples of peptides of the invention include, but are not limited to: DLPAKRGSAPGST (SEQ ID NO: 12), SELPGLKHPCVPGS (SEQ ID NO: 13), TTLGPVKRDSIPGE (SEQ ID NO: 14), SLPIKHDRLPATS (SEQ ID NO: 15), ELPLKRGRVPVE (SEQ ID NO: 16) and NLPDLKKPRVPATS (SEQ ID NO: 17).
[0047] In another embodiment, the present invention provides an isolated linear cationic peptide having an amino acid sequence of the following general formula (general formula C):
X1X2X3X4WX4WX4X5k (SEQ ID NO: 18) (this formula includes CP12a and CP12d), where X1Is one to four amino acids selected from A, P or R, X2Is one or two aromatic amino acids (F, Y and W), X3Is a P or K, X4Is zero to two amino acids selected from A, P, Y or W, X5Is one to three amino acids selected from R or P. Examples of peptides of the invention include, but are not limited to: RPRYPWWPWWPYRPRK (SEQ ID NO: 19), RRAWWKAWWARRK (SEQ ID NO: 20), RAPYWPWAWARPRK (SEQ ID NO: 21), RPAWKYWWPWPWPRRK (SEQ ID NO: 22), RAAFKWAWAWWRRK (SEQ ID NO: 23) and RRRWKWAWPRRK (SEQ ID NO: 24).
[0048] In another embodiment, the present invention provides an isolated hexameric cationic peptide having the amino acid sequence of the following general formula (general formula D):
X1X2X3X4X1VX3X4RGX4X3X4X1X3X1(SEQ ID NO: 25), wherein X1Is one or two of R or K, X2Is a polar or charged amino acid (S, T, M, N, Q, D, E, K, R and H), X3Is C, S, M, D or A, X4Is F, I, V, M or R. Examples of peptides of the invention include, but are not limited to: RRMCIKVCVRGVCRRKCRK (SEQ ID NO: 26), KRSCFKVSMRGVSRRRCK (SEQ ID NO: 27), KKDAIKKVDIRGMDMRRAR (SEQ ID NO: 28), RKMVKVDVRGIMIRKDRR (SEQ ID NO: 29), KQCVKVAMRGMALRRCK (SEQ ID NO: 30) and RREAIRRVAMRGRDMKRMRR (SEQ ID NO: 31).
[0049] In another embodiment, the present invention provides an isolated hexameric cationic peptide having the amino acid sequence of the following general formula (general formula E):
X1X2X3X4X1VX5X4RGX4X5X4X1X3X1(SEQ ID NO: 32) wherein X1Is one or two of R or K, X2Is a polar or charged amino acid (S, T, M, N, Q, D, E, K, R and H), X3Is C, S, M, D or A, X4Is an F, I, V, M or R, X5Is an A, I, S, M, D or R. Examples of peptides of the invention include, but are not limited to: RTCVKRVAMRGIIRKRCR (SEQ ID NO: 33), KKQMMKRVDVRGISVKRKR (SEQ ID NO: 34), KESIKVIIRGMMVRMKK (SEQ ID NO: 35), RRDCRRVMVRGIDIKAK (SEQ ID NO: 36), KRTAIKKVSRRGMSVKARR (SEQ ID NO: 37) and RHCIRRVSMRGIIMRRCK (SEQ ID NO: 38).
[0050] In another embodiment, the invention provides isolated longer cationic peptides having the amino acid sequence of the following general formula (general formula F):
KX1KX2FX2KMLMX2ALKKX3(SEQ ID NO: 39), wherein X1Is a polar amino acid (C, S, T, M, N and Q); x2Is A, L, S or K, X3Is 1-17 amino acids selected from G, A, V, L, I, P, F, S, T, K and H. Examples of peptides of the invention include, but are not limited to: KCKLFKKMLMLALKKVLTTGLPALKLTK (SEQ ID NO: 40), KSKSFLKMLMKALKKVLTTGLPALIS (SEQ ID NO: 41), KTKKFAKMLMMALKKVVSTAKPLAILS (SEQ ID NO: 42), KMKSFAKMLMLALKKVLKVLTTALTLKAGLPS (SEQ ID NO: 43), KNKAFAKMLMKALKKVTTAAKPLTG (SEQ ID NO: 44) and KQKLFAKMLMSALKKKTLVTTPLAGK (SEQ ID NO: 45).
[0051] In another embodiment, the invention provides isolated longer cationic peptides having the amino acid sequence of the following general formula (general formula G):
KWKX2X1X1X2X2X1X2X2X1X1X2X2IFHTALKPISS (SEQ ID NO: 46), wherein X1Is a hydrophobic amino acid, X2Are hydrophilic amino acids. Examples of peptides of the invention include, but are not limited to: KWKSFLRTFKSPVRTIFHTALKPISS (SEQ ID NO: 47), KWKSYAHTIMSPVRLIFHTALKPISS (SEQ ID NO: 48), KWKRGAHRFMKFLSTIFHTALKPISS (SEQ ID NO: 49), KWKKWAHSPRKVLTRIFHTALKPISS (SEQ ID NO: 50), KWKSLVMMFKKPARRIFHTALKPISS (SEQ ID NO: 51), and KWKHALMKAHMLWHMIFHTALKPISS (SEQ ID NO: 52).
[0052] In another embodiment, the invention provides an isolated cationic peptide having an amino acid sequence of the formula:
KWKSFLRTFKSPVRTVFHTALKPISS (SEQ ID NO: 53) or
KWKSYAHTIMSPVRLVFHTALKPISS(SEQ ID NO:54)。
[0053] The term "isolated" as used herein refers to a peptide that is substantially free of other proteins, lipids, and nucleic acids (e.g., cellular components with which the peptide produced in vivo is naturally associated). Preferably, the peptide is at least 70%, 80%, or most preferably 90% pure by weight.
[0054] The invention also includes analogs (analogs), derivatives (derivitives), conservative variants (conservative variations) and cationic peptide variants (cationic peptide variations) of the enumerated polypeptides, provided that the analogs, derivatives, conservative variants or variants have detectable activity in enhancing innate immunity or anti-inflammatory activity. The activity of an analog, derivative, conservative variant or variant of a peptide need not be exactly the same as the activity of the peptide.
[0055] Cationic peptide "variants" refer to peptides having variations of the cationic peptide referred to. For example, the term "variant" includes a cationic peptide in which at least one amino acid of a reference peptide is replaced in an expression library. The term "reference" peptide refers to any cationic peptide of the invention (e.g., as defined in the formulas above) from which variants, derivatives, analogs or conservative variations are derived. The term "derivative" includes hybrid peptides that include at least a portion of each of two cationic peptides (e.g., 30-80% of each of the two cationic peptides). Also included are peptides obtained by deleting one or more amino acids from the sequences of the peptides listed herein, as long as the derivatives have an innate immunity-enhancing or anti-inflammatory activity. Smaller reactive molecules can thus be developed which also have utility. For example, amino-terminal amino acids or carboxy-terminal amino acids that are not essential for enhancing innate immunity and anti-inflammatory activity of the peptide may be removed. Likewise, one or a few (e.g., less than 5) amino acids may be added to a cationic peptide without completely inhibiting the activity of the peptide, thus yielding additional derivatives. In addition, C-terminal derivatives, such as C-terminal methyl esters, and N-terminal derivatives may be obtained and are included in the present invention. The peptides of the present invention include any analog, homolog, mutant, isomer or derivative of the disclosed peptides, as long as the biological activity described herein is retained. Also included are the reverse sequences of the peptides encompassed by the previously proposed formulas. Furthermore, amino acids in the "D" configuration may be replaced by amino acids in the "L" configuration, and vice versa. Alternatively, the peptide may be cyclized by chemical means or by adding two or more cysteine residues to its sequence and oxidizing to form a disulfide bond.
[0056] The invention also includes peptides that are conservative variants of those peptides listed herein. The term "conservative variant" as used herein refers to a polypeptide in which at least one amino acid is replaced with another residue having similar biological activity. Examples of conservative variations include the replacement of one hydrophobic residue, such as isoleucine, valine, leucine, alanine, cysteine, glycine, phenylalanine, proline, tryptophan, tyrosine, norleucine, or methionine for another, or the replacement of one polar residue for another, such as the replacement of arginine for lysine, glutamic for aspartic acids, glutamine for asparagine, and the like. Neutral hydrophilic amino acids that may be substituted for each other include asparagine, glutamine, serine, and threonine. "conservative variants" also include peptides obtained by replacing an unsubstituted parent amino acid(s) with a substituted amino acid. These substituted amino acids may include methylated or amidated amino acids. Other alternatives are well known to those skilled in the art. In one aspect, antibodies made from substituted polypeptides are also capable of specifically binding to unsubstituted polypeptides.
[0057] The peptides of the invention can be synthesized by commonly used methods, for example, which include t-BOC or FMOC protection of the alpha amino group. Both methods involve stepwise synthetic steps in which one amino acid is added to each step starting from the C-terminus of the peptide (see Coligan et al, Current protocols in Immunology, Wiley Interscience, 1991, Unit 9). The peptides of the invention may also be synthesized using well known Solid phase peptide Synthesis methods, such as those described by Merrifield (J.Am.chem.Soc., 85: 2149, 1962) and Stewart and Young (Solid phase peptides Synthesis, Freeman, San Francisco, 1969, pages 27-62) using styrene-divinylbenzene co-polymers with 0.1-1.0 millimoles of amine per gram of polymer. After completion of the chemical synthesis, the peptide was deprotected and cleaved from the polymer by treatment with HF-10% anisole at 0 ℃ for 1/4-1 h. After evaporation of the reagents, the peptides were extracted from the polymer with 1% acetic acid solution and then lyophilized to give the crude product. The peptide is purified using techniques such as gel filtration, for example using 5% acetic acid as solvent on Sephadex G-15. Lyophilization of an appropriate fraction of the column eluate can yield a homogeneous peptide, which can then be characterized by standard techniques such as amino acid analysis, thin layer chromatography, high performance liquid chromatography, ultraviolet absorption spectroscopy, molar rotation, or solubility measurements. If desired, the peptide can be quantified using solid phase Edman degradation.
[0058] The invention also includes isolated nucleic acids (e.g., DNA, cDNA, or RNA) encoding the peptides of the invention. Nucleic acids encoding analogs, mutants, conservative variants, and variants of the peptides described herein are also included. The term "isolated" as used herein refers to nucleic acids that are substantially free of proteins, lipids, and other nucleic acids that are naturally associated with nucleic acids produced in vivo. Preferably, the nucleic acid is at least 70%, 80% or preferably 90% pure by weight. Conventional methods for synthesizing nucleic acids in vitro may be used instead of in vivo methods. As used herein, "nucleic acid" refers to a polymer of deoxyribonucleotides or ribonucleotides, which may be a separate fragment or part of a large genetic construct (construct) (e.g., a promoter linked to a nucleic acid encoding a peptide of the invention). A wide variety of genetic constructs (e.g., plasmids and other expression vectors) are known in the art and can be used to produce the peptides of the invention in cell-free systems or in prokaryotic or eukaryotic (e.g., yeast, insect or mammalian) cells. In view of the degeneracy of the genetic code, a person skilled in the art can readily synthesize a nucleic acid encoding a polypeptide of the invention. The peptides of the invention can be conveniently obtained using conventional molecular biology methods using the nucleic acids of the invention.
[0059] The DNA encoding the cationic peptide of the present invention may be inserted into an "expression vector". The term "expression vector" refers to a genetic construct such as a plasmid, virus, or other vector known in the art that can be designed to contain a nucleic acid encoding a polypeptide of the present invention. These expression vectors are preferably plasmids containing promoter sequences which facilitate transcription of the inserted gene sequences in the host cell. Expression vectors typically contain an origin of replication, a promoter, and a polynucleotide (e.g., an antibiotic resistance polynucleotide) that enables phenotypic selection of the transformed cell. Various promoters can be used in the present invention, including inducible and constitutive promoters. Typically, the expression vector contains a replicon site and control sequences derived from a species compatible with the host cell.
[0060]The nucleic acids of the invention may be transformed or transfected into a recipient (recipient) using conventional techniques well known to those skilled in the art. For example, when the host cell is Escherichia coli (E.coli), CaCl known in the art may be used2、MgCl2Or the RbCl method to prepare competent cells capable of taking up DNA. Alternatively, physical methods such as electroporation or microinjection may be used. Electroporation is the transfer of nucleic acids into cells by high voltage pulses. In addition, the nucleic acid can be introduced into the host cell by protoplast fusion using methods well known in the art. Suitable methods for transforming eukaryotic cells, such as electroporation and lipofection, are also known.
[0061] The invention encompasses "host cell" or "recipient cell" refers to any cell in which a polypeptide of the invention can be expressed using a nucleic acid of the invention. The term also includes any progeny of the recipient cell or host cell. Preferred recipient cells or host cells of the invention include e.coli (e.coli), staphylococcus aureus (s.aureus) and pseudomonas aeruginosa (p.aeruginosa), although other gram-negative and gram-positive bacteria, fungi and mammalian cells and other organisms known in the art may also be utilized, provided that the expression vector contains an origin of replication to allow expression in the host.
[0062] The cationic peptide polynucleotide sequences used in accordance with the methods of the present invention can be isolated from an organism or synthesized in the laboratory. A specific DNA sequence encoding a cationic peptide of interest can be obtained by: 1) isolating double-stranded DNA sequences from the genomic DNA; 2) chemically synthesizing a DNA sequence to provide codons required for the cationic peptide of interest; and 3) in vitro synthesis of double stranded DNA sequences using reverse transcription of mRNA isolated from donor cells. In the latter case, the complementary sequence of the mRNA, which is usually referred to as cDNA, is obtained as double-stranded DNA.
[0063] When the entire sequence of amino acid residues of the desired peptide product is known, synthesis of its DNA sequence is often the method of choice. In the present invention, the synthetic DNA sequence has an advantage of allowing incorporation of codons most likely to be recognized by a bacterial host, thereby allowing high-level expression without translational difficulty. In addition, virtually any peptide can be synthesized, including those that encode natural cationic peptides, variants thereof, or synthetic peptides.
[0064] When the entire sequence of the desired peptide is unknown, direct synthesis of the DNA sequence is not possible, and in this case the method of choice is to obtain the cDNA sequence. Standard procedures for isolating cDNA sequences of interest include constructing plasmids or phages containing cDNA libraries obtained by reverse transcription of large amounts of mRNA present in donor cells with high levels of genetic expression. When used with the polymerase chain reaction, even rare expression products can be cloned. When a significant portion of the amino acid sequence of the cationic peptide is known, labelled single or double stranded DNA or RNA probes can be made whose sequence is believed to be present in the cDNA of interest, and these probes can be used in DNA/DNA hybridization experiments on copies of cDNA clones which have been denatured to single stranded form (Jay et al, Nuc.acid.Res., 11: 2325, 1983).
[0065] The peptides of the invention may be administered parenterally by injection or by gradual infusion over a period of time. The peptide may be administered intravenously, intraperitoneally, intramuscularly, subcutaneously, intracavity, or transdermally. Preferred methods of delivery of the peptide include oral delivery via microspheres or protein-like capsules, delivery to the lungs in aerosol form, or transdermal delivery using iontophoresis or electroporation. Other methods of administration are known to those skilled in the art.
[0066] The preparation of parenteral administration forms of the peptides of the invention includes sterile aqueous or anhydrous solutions, suspensions and emulsions. Examples of anhydrous solvents are propylene glycol, polyethylene glycol, vegetable oils such as olive oil, and injectable organic esters such as ethyl oleate. Aqueous carriers include water, ethanol/water solutions, emulsions or suspensions, including saline and parenteral vehicles containing buffer media, including sodium chloride solutions, ringer's dextrose solution, dextrose and sodium chloride, sodium acetate, sodium citrate, lactated ringer's solution or non-volatile oils. Carriers for intravenous administration include fluid and nutritional supplements, electrolyte supplements (e.g., they may be based on ringer's dextrose solution), and the like. Preservatives and other additives may also be present such as, for example, antimicrobial agents, antioxidants, chelating agents, inert gases and the like.
[0067] The invention will now be described in more detail by reference to the following non-limiting examples. The invention has been described in greater detail by reference to certain preferred embodiments thereof, and it is to be understood that modifications and variations are within the spirit and scope of the disclosure as described and claimed.
Example 1
Anti-sepsis/anti-inflammatory activity
[0068]A polynucleotide array was used to determine the effect of cationic peptides on the transcriptional response of epithelial cells. The a549 human epithelial cell line was maintained in dmem (gibco) and supplemented with 10% fetal bovine serum (FBS, Medicorp). A549 cells were plated in 100mm tissue culture dishes, one for each cultureThe culture dish contains 2.5 × 106Cells, cultured overnight, then mixed with 100ng/ml e.colio111: b4LPS (Sigma) was incubated for 4 hours with 50. mu.g/ml peptide and medium or without peptide and medium alone as a control. After stimulation, cells were washed once with diethylpyrocarbonate-treated Phosphate Buffered Saline (PBS) and scraped with a cell brush. Total RNA was isolated using RNAquesous (Ambion, Austin, TX). The RNA pellet was resuspended In RNase-free water containing Superase-In (RNase inhibitor; Ambion). DNA contamination was removed using a DNA-free kit (Ambion). The quality of the RNA was estimated by electrophoresis on a 1% agarose gel.
[0069] The polynucleotide array used was a Human Operon array (identification number of the genome is PRHU04-S1) consisting of duplicate 14,000 Human oligomer spots. Probes were prepared with 10. mu.g total RNA and labeled with Cy3 or Cy5 labeled dUTP. The probe was purified and hybridized to a printed glass slide, overnight at 42 ℃ and then washed. After washing, images were taken with a Perkin Elmer array scanner. The mean, median and background intensities of the spots were determined using image processing software (Imapolynucheotide 5.0, Marina Del Rey, Calif.). The background was removed using a "homemade" procedure. The program calculates the bottom intensity of each sub-cell (subgrid) as 10% and subtracts this value for each cell (grid). The analysis was performed using Genespring software (Redwood City, Calif.). The intensity value of the middle spot is obtained from a collection of spot values within a slide and compared to the values of all slides in this experiment, thereby normalizing the intensity of each spot. The relative change between the peptide-treated cells and the control cells can be seen in tables 1 and 2. Table 2 shows only those polynucleotides whose expression varies significantly among the 14,000 polynucleotides tested. This data indicates that the peptide has the ability to broadly reduce LPS-induced polynucleotide expression.
[0070] In table 1, a study of the polynucleotide microarray indicated that SEQ ID NO: 27 effectively reduced the expression of c.coli O111: b4 expression of various polynucleotides upregulated by LPS. The peptides (50. mu.g/ml) and LPS (0.1. mu.g/ml) were incubated with A549 cells for 4 hours, or LPS alone was incubated with A549 cells for 4 hours, followed by isolation of RNA. Cy3/Cy 5-labeled cDNA probes were prepared from 5. mu.g total RNA and hybridized to a HumanOperon array (PRHU 04). The third column of table 1 shows the intensity of the unstimulated cells. "ratio: LPS/control "column refers to the result of dividing the intensity of polynucleotide expression in cells stimulated with LPS by the intensity of non-stimulated cells. "ratio: LPS + ID 27/control "column refers to the intensity of polynucleotide expression in cells stimulated with LPS and peptide divided by the intensity of unstimulated cells.
Table 1: peptide SEQ ID27 reduction
Coli O111: b4 LPS-upregulated a549 human epithelial cell polynucleotide expression
| Sequence accession numbera | Polynucleotide Gene function | Comparison: medium strength only | The ratio is: LPS/control | The ratio is: LPS + ID 27/control |
| AL031983 | Is unknown | 0.032 | 302.8 | 5.1 |
| L04510 | ADP-ribosylation factor | 0.655 | 213.6 | 1.4 |
| D87451 | Ring finger protein 10 | 3.896 | 183.7 | 2.1 |
| AK000869 | Hypothetical proteins | 0.138 | 120.1 | 2.3 |
| U78166 | Ric-like expression in neurons | 0.051 | 91.7 | 0.2 |
| AJ001403 | Tracheal bronchus mucin 5 subtype B | 0.203 | 53.4 | 15.9 |
| AB040057 | Serine/threonine protein kinase MASK | 0.95 | 44.3 | 15.8 |
| Z99756 | Is unknown | 0.141 | 35.9 | 14.0 |
| L42243 | Interferon receptor 2 | 0.163 | 27.6 | 5.2 |
| MN_016216 | RNA lasso debranching enzyme | 6.151 | 22.3 | 10.9 |
| AK001589 | Hypothetical proteins | 0.646 | 19.2 | 1.3 |
| AL137376 | Is unknown | 1.881 | 17.3 | 0.6 |
| AB007856 | FEM-1-like death receptor binding proteins | 2.627 | 15.7 | 0.6 |
| AB007854 | Growth arrest specific protein 7 | 0.845 | 14.8 | 2.2 |
| AK000353 | Cytosolic ovarian tumor antigen 1 | 0.453 | 13.5 | 1.0 |
| D14539 | Myeloid/lymphoid or mixed lineage leukemia; is translocated; 1(MLLT1) | 2.033 | 11.6 | 3.1 |
| X76785 | Integration site of epstein-barr virus | 0.728 | 11.6 | 1.9 |
| M54915 | Pim-1 oncogene | 1.404 | 11.4 | 0.6 |
| NM_006092 | Caspase recruitment domain 4 | 0.369 | 11.0 | 0.5 |
| J03925 | Integrin alpha M | 0.272 | 9.9 | 4.2 |
| NM_001663 | ADP-ribosylation factor 6 | 0.439 | 9.7 | 1.7 |
| M23379 | RAS p21 protein activator | 0.567 | 9.3 | 2.8 |
| K02581 | Soluble thymidine kinase 1 | 3.099 | 8.6 | 3.5 |
| U94831 | Transmembrane 9 superfamily member 1 | 3.265 | 7.1 | 1.5 |
| X70394 | Zinc finger protein 146 | 1.463 | 6.9 | 1.7 |
| AL137614 | Hypothetical proteins | 0.705 | 6.8 | 1.0 |
| U43083 | Guanine nucleotide binding proteins | 0.841 | 6.6 | 1.6 |
| AL137648 | DKFZp434J1813 protein | 1.276 | 6.5 | 0.8 |
| AF085692 | ATP-binding cassette subfamily C (CFTR-MRP) member 3 | 3.175 | 6.5 | 2.4 |
| AK001239 | Hypothetical protein FLJ10377 | 2.204 | 6.4 | 1.3 |
| NM_001679 | ATP enzyme Na+/K+Transporter beta 3 polypeptides | 2.402 | 6.3 | 0.9 |
| L24804 | Inactive progesterone receptors | 3.403 | 6.1 | 1.1 |
| U15932 | Dual specific phosphatase 5 | 0.854 | 6.1 | 2.1 |
| M36067 | ATP-dependent DNA ligase I | 1.354 | 6.1 | 2.2 |
| AL161951 | Is unknown | 0.728 | 5.8 | 1.9 |
| M59820 | Colony stimulating factor 3 receptor | 0.38 | 5.7 | 2.0 |
| AL050290 | Spermidine/spermine N1-acyltransferase | 2.724 | 5.6 | 1.4 |
| NM_002291 | Laminin beta-1 | 1.278 | 5.6 | 1.8 |
| X06614 | Retinoic acid receptor alpha | 1.924 | 5.5 | 0.8 |
| AB007896 | Putative L-form neutral ammoniaAmino acid transporters | 0.94 | 5.3 | 1.8 |
| AL050333 | DKFZP564B116 protein | 1.272 | 5.3 | 0.6 |
| AK001093 | Hypothetical proteins | 1.729 | 5.3 | 2.0 |
| NM_016406 | Hypothetical proteins | 1.314 | 5.2 | 1.2 |
| M86546 | Pre-B cell leukemia transcription factor 1 | 1.113 | 5.2 | 2.2 |
| X56777 | Zona pellucida glycoprotein 3A | 1.414 | 5.0 | 1.4 |
| NM_013400 | Replication initiation region protein | 1.241 | 4.9 | 2.0 |
| NM_002309 | Leukemia inhibitory factor | 1.286 | 4.8 | 1.9 |
| NM_001940 | Dentate red nucleus globus pallidus atrophy | 2.034 | 4.7 | 1.2 |
| U91316 | Cytosolic acyl-CoA thioester hydrolase | 2.043 | 4.7 | 1.4 |
| X76104 | Death-related protein kinase 1 | 1.118 | 4.6 | 1.8 |
| AF131838 | Is unknown | 1.879 | 4.6 | 1.4 |
| AL050348 | Is unknown | 8.502 | 4.4 | 1.7 |
| D42085 | KIAA0095 gene product | 1.323 | 4.4 | 1.2 |
| X92896 | Is unknown | 1.675 | 4.3 | 1.5 |
| U26648 | Syntaxin 5A | 1.59 | 4.3 | 1.4 |
| X85750 | Is associated with the differentiation of monocytes into macrophages | 1.01 | 4.3 | 1.1 |
| D14043 | CD164 antigen sialoglycoproteins | 1.683 | 4.2 | 1.0 |
| J04513 | Fibroblast growth factor 2 | 1.281 | 4.0 | 0.9 |
| U19796 | Melanoma associated antigens | 1.618 | 4.0 | 0.6 |
| AK000087 | Hypothetical proteins | 1.459 | 3.9 | 1.0 |
| AK001569 | Hypothetical proteins | 1.508 | 3.9 | 1.2 |
| AF189009 | Ubiquitin 2 | 1.448 | 3.8 | 1.3 |
| U60205 | sterol-C4-methyl oxidase-like | 1.569 | 3.7 | 0.8 |
| AK000562 | Hypothetical proteins | 1.166 | 3.7 | 0.6 |
| AL096739 | Is unknown | 3.66 | 3.7 | 0.5 |
| AK000366 | Hypothetical proteins | 15.192 | 3.5 | 1.0 |
| NM_006325 | RAN Member RAS oncogene superfamily | 1.242 | 3.5 | 1.4 |
| X51688 | Cyclin A2 | 1.772 | 3.3 | 1.0 |
| U34252 | Aldehyde dehydrogenase | 1.264 | 3.3 | 1.2 |
| NM_013241 | Protein containing FH1/FH2 structural domain | 1.264 | 3.3 | 0.6 |
| AF112219 | Esterase/formyl glutathione hydrolase | 1.839 | 3.3 | 1.1 |
| NM_016237 | Interphase promoting complex subunit 5 | 2.71 | 3.2 | 0.9 |
| AB014569 | KIAA0669 Gene product | 2.762 | 3.2 | 0.2 |
| AF151047 | Hypothetical proteins | 3.062 | 3.1 | 1.0 |
| X92972 | Protein phosphatase 6 catalytic subunit | 2.615 | 3.1 | 1.1 |
| AF035309 | Proteasome 26S subunit ATPase 5 | 5.628 | 3.1 | 1.3 |
| U52960 | SRB7 homologs | 1.391 | 3.1 | 0.8 |
| J04058 | Electron transfer flavoprotein alpha polypeptide | 3.265 | 3.1 | 1.2 |
| M57230 | Interleukin 6 signal transducer | 0.793 | 3.1 | 1.0 |
| U78027 | Alpha galactosidase enzyme | 3.519 | 3.1 | 1.1 |
| AK000264 | Is unknown | 2.533 | 3.0 | 0.6 |
| X80692 | Mitogen activated protein kinase 6 | 2.463 | 2.9 | 1.3 |
| L25931 | Nuclear lamin B receptor | 2.186 | 2.7 | 0.7 |
| X13334 | CD14 antigen | 0.393 | 2.5 | 1.1 |
| M32315 | Tumor necrosis factor receptor superfamily member 1B | 0.639 | 2.4 | 0.4 |
| NM_004862 | TNF-alpha factor induced by LPS | 6.077 | 2.3 | 1.1 |
| AL050337 | Interferon gamma receptor 1 | 2.064 | 2.1 | 1.0 |
aAll accession numbers in tables 1 through 64 refer to GenBank accession numbers (accession numbers).
[0071] In table 2, polynucleotide microarray studies showed that cationic peptide at a concentration of 50 μ g/ml effectively reduced the level of peptide expressed by e.coli O111 at 100 ng/ml: b4 expression of a plurality of polynucleotides upregulated by LPS. The peptides and LPS were incubated with a549 cells for 4 hours, or LPS alone was incubated with a549 cells for 4 hours, followed by isolation of RNA. Cy3/Cy 5-labeled cDNA probes were prepared using 5. mu.g total RNA and hybridized to a Human Operon array (PRHU 04). The third column of table 2 shows the intensity of the unstimulated cells. "ratio: LPS/control "column refers to the result of dividing the intensity of polynucleotide expression in cells stimulated with LPS by the intensity of non-stimulated cells. The other columns refer to the results of dividing the intensity of polynucleotide expression in cells stimulated with LPS and peptide by the intensity of unstimulated cells.
[0072] Table 2: coli O111: b4LPS upregulated and decreased human A549 epithelial cell polynucleotide expression by cationic peptides
| Registration number | Gene | Comparison: medium strength only | The ratio is: LPS/control | The ratio is: LPS + ID 27/control | The ratio is: LPS + ID 16/control | The ratio is: LPS + ID 22/control |
| AL031983 | Is unknown | 0.03 | 302.8 | 5.06 | 6.91 | 0.31 |
| L04510 | ADP-ribosylation factor | 0.66 | 213.6 | 1.4 | 2.44 | 3.79 |
| D87451 | Ring finger protein | 3.90 | 183.7 | 2.1 | 3.68 | 4.28 |
| AK000869 | Hypothetical proteins | 0.14 | 120.1 | 2.34 | 2.57 | 2.58 |
| U78166 | Ric sample | 0.05 | 91.7 | 0.20 | 16.88 | 21.37 |
| X03066 | Class II MHCDO beta | 0.06 | 36.5 | 4.90 | 12.13 | 0.98 |
| AK001904 | Hypothetical proteins | 0.03 | 32.8 | 5.93 | 0.37 | 0.37 |
| AB037722 | Is unknown | 0.03 | 21.4 | 0.30 | 0.30 | 2.36 |
| AK001589 | Hypothetical proteins | 0.65 | 19.2 | 1.26 | 0.02 | 0.43 |
| AL137376 | Is unknown | 1.88 | 17.3 | 0.64 | 1.30 | 1.35 |
| L19185 | Thioredoxin dependent peroxiredoxin reductase 1 | 0.06 | 16.3 | 0.18 | 2.15 | 0.18 |
| J05068 | Transcobalamin protein I | 0.04 | 15.9 | 1.78 | 4.34 | 0.83 |
| AB007856 | FEM-1-like death receptor binding | 2.63 | 15.7 | 0.62 | 3.38 | 0.96 |
| Protein | ||||||
| Ak000353 | Cytosolic ovarian tumor antigen 1 | 0.45 | 13.5 | 1.02 | 1.73 | 2.33 |
| X16940 | Smooth muscle intestinal actin gamma 2 | 0.21 | 11.8 | 3.24 | 0.05 | 2.26 |
| M54915 | Pim-1 oncogene | 1.40 | 11.4 | 0.63 | 1.25 | 1.83 |
| AL122111 | Hypothetical proteins | 0.37 | 10.9 | 0.21 | 1.35 | 0.03 |
| M95678 | Phospholipase C beta 2 | 0.22 | 7.2 | 2.38 | 0.05 | 1.33 |
| AK001239 | Hypothetical proteins | 2.20 | 6.4 | 1.27 | 1.89 | 2.25 |
| AC004849 | Is unknown | 0.14 | 6.3 | 0.07 | 2.70 | 0.07 |
| X06614 | Retinoic acid receptor alpha | 1.92 | 5.5 | 0.77 | 1.43 | 1.03 |
| AB007896 | Putative L-type neutral amino acid transporter | 0.94 | 5.3 | 1.82 | 2.15 | 2.41 |
| AB010894 | BAI1 related protein | 0.69 | 5.0 | 1.38 | 1.03 | 1.80 |
| U52522 | RAC1 mate | 1.98 | 2.9 | 1.35 | 0.48 | 1.38 |
| AK001440 | Hypothetical proteins | 1.02 | 2.7 | 0.43 | 1.20 | 0.01 |
| NM_001148 | Ankyrin 2 of neurons | 0.26 | 2.5 | 0.82 | 0.04 | 0.66 |
| X07173 | Alpha inhibitor H2 | 0.33 | 2.2 | 0.44 | 0.03 | 0.51 |
| AF095687 | Brain and nasopharyngeal carcinoma sensitive protein | 0.39 | 2.1 | 0.48 | 0.03 | 0.98 |
| NM_016382 | NK cell activation-inducing ligand NAIL | 0.27 | 2.1 | 0.81 | 0.59 | 0.04 |
| AB023198 | KIAA0981 protein | 0.39 | 2.0 | 0.43 | 0.81 | 0.92 |
Example 2
Neutralizing stimulation of immune cells
[0073]The ability of the compounds to suppress the stimulation of immune cells by gram-negative bacteria and gram-positive bacterial products was tested. Bacterial products stimulate cells of the immune system, thereby producing inflammatory cytokines, which when uncompressed, can lead to sepsis. The experiments were initiated using the murine macrophage line RAW264.7, human epithelial cell line A549 and primary macrophages from BALB/c murine bone marrow obtained from the American type culture Collection (Manassas, Va.), and primary macrophages from Charles River Laboratories, Wilmington, Mass.). Cells from murine bone marrow were cultured in 150mm culture plates in Darbeck modified eagle's medium (DMEM; Life Technologies, Burlington, ON) supplemented with 20% FBS (Sigma chemical Co, St. Louis, Mo.) and 20% L of cell conditioned medium as the M-CSF source. Removing the culture medium with L cell conditioned medium when macrophage cells are 60-80% full,incubate for 14-16 hours until they go to quiescent state, then treat with 10ng/ml LPS or 100ng/ml LPS + 20. mu.g/ml peptide for 24 hours. By ELISA (R)&D Systems, Minneapolis, MN) determined the cytokine release into the culture supernatant. Cell lines RAW264.7 and a549 were maintained in DMEM supplemented with 10% fetal calf serum. RAW264.7 cells at 10 per well6The density of individual cells was seeded in 24-well DMEM-containing plates, and A549 cells were plated at 10 per well5Individual cell densities were seeded into 24-well plates containing DMEM, both at 37 ℃ in 5% CO2Culturing overnight. DMEM was aspirated from overnight grown cells and replaced with fresh medium. In some experiments, volunteers' blood was collected by venipuncture (following procedures approved by the ethical committee for UBC clinical studies, certification No. C00-0537) into tubes containing 14.3USP units heparin/ml blood (Becton Dickinson, Franklin Lakes, NJ). The blood was mixed with LPS with or without peptide in a polypropylene tube at 37 ℃ for 6 hours. Samples were centrifuged at 2000 Xg for 5 min, plasma collected and stored at-20 ℃ until ELISA (R)&D Systems) was taken out for IL-8 analysis. In these cell experiments, cells were incubated with LPS or other bacterial products at 37 ℃ in 5% CO2Incubated for 6-24 hours. Salmonella typhimurium LPS and e.coli 0111: b4LPS was purchased from Sigma. Lipoteichoic acid (LTA) from Staphylococcus aureus (Sigma) was resuspended in endotoxin free water (Sigma). The LTA preparation was subjected to the limulus amebocyte lysate test (Sigma) to confirm that it was not significantly contaminated with endotoxin. Endotoxin contamination was less than 1ng/ml and this concentration did not result in significant cytokine production by RAW264.7 cells. Capless lipoarabinomannan (AraLAM) was gifted by doctor John t. belisle, Colorado State University. The AraLAM of Mycobacterium (Mycobacterium) was filter sterilized and endotoxin contamination was determined by limulus amoebocyte assay and found to be 3.75ng/1.0mg LAM. A range of concentrations of cationic peptide is added simultaneously with (or subsequently to) the addition of LPS. The supernatant was removed and ELISA (R) was used&D Systems) were tested for the production of cytokines. All experiments were performed at least three times and similar results were obtained. To prove thatAnti-sepsis activity in entities, 2 or 3 μ g e.coli 0111: b4LPS phosphate buffered saline (PBS; pH 7.2) was injected intraperitoneally into galactosamine sensitized female CD-1 or BALB/c mice for 8-10 weeks, thereby inducing sepsis. In other experiments, 400 μ g e.coli 0111: b4LPS was injected into CD-1 mice 10 minutes later, and the peptide (200. mu.g) was introduced by intraperitoneal injection. Survival was monitored 48 hours after injection.
[0074] It has been traditionally thought that the production of excess TNF- α is linked to the onset of sepsis. The three types of LPS, LTA or AraLAM used in this example represent products released by gram-negative and gram-positive bacteria. SEQ ID NO: 1 is capable of significantly reducing the level of a peptide derived from salmonella typhimurium, burkholderia cepacia (b.cepacia) and e.coli0111: b4 TNF- α production stimulated by LPS was slightly less affected (Table 3). In the latter two cases it can be seen that peptides at concentrations as low as 1. mu.g/ml (0.25nM) can also lead to a significant reduction in TNF-. alpha.production. A different peptide, SEQ ID NO: 3 did not reduce LPS-induced TNF- α production in RAW macrophages, suggesting that this is a non-uniform and unpredictable property of the cationic peptide. Representative peptides of each formula were also tested for their effect as measured by e.coli 0111: b4 ability of LPS to stimulate TNF- α production (table 4). Although many of these peptides reduced TNF- α production by at least 60%, their ability to reduce TNF- α production was differential.
[0075] Peptides SEQ ID NO: 1 and SEQ ID NO: 2 can also impair the ability of the bacterial product to stimulate the production of IL-8 by epithelial cell lines. LPS is known to be effective in stimulating IL-8 production by epithelial cells. The peptides were able to suppress the IL-8 production response of epithelial cells to LPS at low concentrations (1-20. mu.g/ml) (tables 5-7). The peptide SEQ ID2 also inhibited IL-8 production in human whole blood induced by LPS (Table 4). In contrast, high concentrations of the peptide SEQ ID NO: 1 (50-100. mu.g/ml) actually resulted in elevated IL-8 levels (Table 5). This indicates that the peptides have different effects at different concentrations.
[0076] The effect of the peptide on inflammatory stimulation was also confirmed in primary murine cells, the peptide SEQ id no: 1 significantly reduced TNF- α production (> 90%) in bone marrow derived macrophages of BALB/c mice, which had been treated with 100ng/ml e.coli 0111: b4 was LPS-stimulated (table 8). These experiments were performed in the presence of serum, which contains the LPS Binding Protein (LBP), a protein that mediates the rapid binding of LPS to CD 14. Coli LPS at 100ng/ml one hour after stimulation with SEQ ID NO: 1 delayed addition to the macrophage supernatant still resulted in a significant reduction in TNF-. alpha.production (70%, Table 9).
[0077] SEQ ID NO: 1 are capable of preventing the in vitro induction of TNF- α by LPS, and in accordance with this, certain peptides are also capable of protecting mice from lethal shock induced by high concentrations of LPS. In some experiments, CD-1 mice were made allergic to LPS by pre-injection of galactosamine. Mice sensitized with galactosamine were injected with 3 μ g of e.coli 0111: b4LPS was sacrificed within 4-6 hours thereafter. At 15 minutes after LPS injection, 200 μ g of SEQ ID NO: 1, 50% of the mice survived (Table 10). In other experiments, higher concentrations of LPS were injected into BALB/c mice without prior injections of galactosamine type D, and the protection provided by the peptide was 100%, compared to no survival in the control group (table 13). The other peptides selected were also found to be protective in these models (tables 11, 12).
[0078] Cationic peptides can also slow down the stimulation of macrophages by products of gram-positive bacteria, such as hated-lipid arabinomannan (AraLAM) of mycobacteria and LTA of staphylococcus aureus (s. For example, SEQ ID NO: 1 inhibited the induction of TNF-. alpha.in RAW264.7 cells by the gram-positive bacterial products LTA (Table 14) and AraLAM (Table 15), to a lesser extent. Another peptide, SEQ ID NO: 2 reduced LTA induction of TNF- α in RAW264.7 cells. 1 μ g/ml of SEQ ID NO: 1 was able to significantly reduce (> 75%) the induction of TNF-. alpha.production by Staphylococcus aureus LTA at 1. mu.g/ml. In SEQ ID NO: 1 concentration of 20 mug/ml, the inhibition rate of AraLAM induced TNF-alpha is more than 60%. Polymyxin b (pmb) can be introduced as a control to demonstrate that the amino acid sequence shown in SEQ ID NO: 1 inhibition of AraLAM induced TNF- α endotoxin contamination was not a significant factor. These results demonstrate that cationic peptides can attenuate the pro-inflammatory cytokine response of the immune system to bacterial products.
[0079] Table 3: in RAW264.7 cells SEQ ID NO: 1 reduces TNF- α production induced by LPS. In the presence of the indicated concentrations of SEQ ID1, the total volume of the cells was measured using 100ng/ml Salmonella typhimurium LPS, 100ng/ml Burkholderia cepacia LPS and 100ng/ml E.coli0111: b4LPS, stimulated RAW264.7 murine macrophages for 6 hours. The concentration of TNF-. alpha.released into the culture supernatant was determined by ELISA. 100% represents the amount of TNF- α obtained by incubating RAW264.7 cells with LPS alone for 6 hours (salmonella typhimurium LPS ═ 34.5 ± 3.2ng/ml, burkholderia cepacia LPS ═ 11.6 ± 2.9ng/ml, e.coli 0111: B4LPS ═ 30.8 ± 2.4 ng/ml). Background levels of TNF- α production from RAW264.7 cells cultured for 6 hours without stimulation were 0.037-0.192 ng/ml. Experimental data were derived from two identical samples and are expressed as the mean + standard deviation of three experiments.
| Amount of SEQ ID1 (. mu.g/ml) | Inhibition of TNF-alpha (%)* | ||
| Burkholderia cepacia LPS | Escherichia coli LPS | Salmonella typhimurium LPS | |
| 0.1 | 8.5±2.9 | 0.0±0.6 | 0.0±0 |
| 1 | 23.0±11.4 | 36.6±7.5 | 9.8±6.6 |
| 5 | 55.4±8 | 65.0±3.6 | 31.1±7.0 |
| 10 | 63.1±8 | 75.0±3.4 | 37.4±7.5 |
| 20 | 71.7±5.8 | 81.0±3.5 | 58.5±10.5 |
| 50 | 86.7±4.3 | 92.6±2.5 | 73.1±9.1 |
[0080] Table 4: cationic peptides reduced TNF- α production induced by e.coli LPS in RAW264.7 cells. In the presence of the specified concentration of cationic peptide, the concentration of the peptide was measured with a 100ng/ml e.coli0111: b4LPS stimulated RAW264.7 murine macrophages for 6 hours. The concentration of TNF-. alpha.released into the culture supernatant was determined by ELISA. Background levels of TNF- α production from RAW264.7 cells cultured for 6 hours without stimulation were 0.037-0.192 ng/ml. Experimental data were derived from two identical samples and are expressed as the mean + standard deviation of three experiments.
| Peptide (20. mu.g/ml) | Inhibition of TNF-alpha (%) |
| SEQ ID 5 | 65.6±1.6 |
| SEQ ID 6 | 59.8±1.2 |
| SEQ ID 7 | 50.6±0.6 |
| SEQ ID 8 | 39.3±1.9 |
| SEQ ID 9 | 58.7±0.8 |
| SEQ ID 10 | 55.5±0.52 |
| SEQ ID 12 | 52.1±0.38 |
| SEQ ID 13 | 62.4±0.85 |
| SEQ ID 14 | 50.8±1.67 |
| SEQ ID 15 | 69.4±0.84 |
| SEQ ID 16 | 37.5±0.66 |
| SEQ ID 17 | 28.3±3.71 |
| SEQ ID 19 | 69.9±0.09 |
| SEQ ID 20 | 66.1±0.78 |
| SEQ ID 21 | 67.8±0.6 |
| SEQ ID 22 | 73.3±0.36 |
| SEQ ID 23 | 83.6±0.32 |
| SEQ ID 24 | 60.5±0.17 |
| SEQ ID 26 | 54.9±1.6 |
| SEQ ID 27 | 51.1±2.8 |
| SEQ ID 28 | 56±1.1 |
| SEQ ID 29 | 58.9±0.005 |
| SEQ ID 31 | 60.3±0.6 |
| SEQ ID 33 | 62.1±0.08 |
| SEQ ID 34 | 53.3±0.9 |
| SEQ ID 35 | 60.7±0.76 |
| SEQ ID 36 | 63±0.24 |
| SEQ ID 37 | 58.9±0.67 |
| SEQ ID 38 | 54±1 |
| SEQ ID 40 | 75±0.45 |
| SEQ ID 41 | 86±0.37 |
| SEQ ID 42 | 80.5±0.76 |
| SEQ ID 43 | 88.2±0.65 |
| SEQ ID 44 | 44.9±1.5 |
| SEQ ID 45 | 44.7±0.39 |
| SEQ ID 47 | 36.9±2.2 |
| SEQ ID 48 | 64±0.67 |
| SEQ ID 49 | 86.9±0.69 |
| SEQ ID 53 | 46.5±1.3 |
| SEQ ID 54 | 64±0.73 |
[0081]Table 5: SEQ ID1 reduced IL-8 production induced by LPS in a549 cells. A549 cells were stimulated with increasing concentrations of SEQ ID1 for 24 hours in the presence of LPS (100ng/ml e.coli 0111: B4). IL-8 concentration in the cultures was determined by ELISA. The background level of IL-8 in individual cells was 0.172. + -. 0.029 ng/ml. Data are presented as mean + standard deviation of three experiments.
| SEQ ID 1(μg/ml) | Inhibition of IL-8 (%) |
| 0.1 | 1±0.3 |
| 1 | 32±10 |
| 10 | 60±9 |
| 20 | 47±12 |
| 50 | 40±13 |
| 100 | 0 |
[0082] Table 6: SEQ ID2 reduced IL-8 production induced by e.coli LPS in a549 cells. Human a549 epithelial cells were stimulated with increasing concentrations of SEQ ID2 for 24 hours in the presence of LPS (100ng/ml e. The IL-8 concentration in the culture supernatant was determined by ELISA. Data are presented as mean + standard deviation of three experiments.
| Concentration of SEQ ID2 (. mu.g/ml) | Inhibition of IL-8 (%) |
| 0.1 | 6.8±9.6 |
| 1 | 12.8±24.5 |
| 10 | 29.0±26.0 |
| 50 | 39.8±1.6 |
| 100 | 45.0±3.5 |
[0083] Table 7: SEQ ID2 reduces IL-8 production induced by e. Coli O111: b4LPS and increasing concentrations of peptide stimulated human whole blood for 4 hours. Human whole blood samples were centrifuged, serum removed and IL-8 concentration determined by ELISA. Data are presented as the mean of two donors.
| SEQ ID 2(μg/ml) | IL-8(pg/ml) |
| 0 | 3205 |
| 10 | 1912 |
| 50 | 1458 |
[0084] Table 8: SEQ ID1 reduces TNF- α production induced by e. BALB/c murine bone marrow-derived macrophages were mixed with 100ng/ml E.coli O111: b4LPS was co-cultured for 6 hours or 24 hours. Supernatants were collected and TNF-. alpha.levels were determined by ELISA. Data represent the amount of TNF-. alpha.that was obtained from two identical experiments with bone marrow-derived macrophages incubated with LPS alone for 6 hours (1.1. + -. 0.09ng/ml) or 24 hours (1.7. + -. 0.2 ng/ml). Background levels of TNF- α were: the concentration was 0.038. + -. 0.008ng/ml at 6 hours, and 0.06. + -. 0.012ng/ml at 24 hours.
| SEQ ID 1(μg/ml) | Production amount (ng/ml) of TNF-. alpha. | |
| 6 hours | 24 hours | |
| LPS Only | 1.1 | 1.7 |
| 1 | 0.02 | 0.048 |
| 10 | 0.036 | 0.08 |
| 100 | 0.033 | 0.044 |
| Control without LPS | 0.038 | 0.06 |
[0085] Table 9: postpone the addition of SEQ ID1 to a549 cells from inhibiting TNF- α production induced by e. At gradually later time points, peptides (20 μ g/ml) were added to the cells already containing a549 human epithelial cells and 100ng/ml e.coli O111: b4LPS culture wells. Supernatants were collected after 6 hours and TNF-. alpha.levels were determined by ELISA. Data are presented as mean + standard deviation of three experiments.
| Time to addition of SEQ ID1 after LPS addition (min) | Inhibition of TNF-alpha (%) |
| 0 | 98.3±0.3 |
| 15 | 89.3±3.8 |
| 30 | 83±4.6 |
| 60 | 68±8 |
| 90 | 53±8 |
[0086] Table 10: the protection against fatal endotoxemia in the semi-lactosamine sensitized CD-1 mice is performed by SEQ ID 1. CD-1 mice (9 weeks old) were made allergic to endotoxin by three intraperitoneal injections of galactosamine (20mg, dissolved in 0.1ml sterile PBS). Coli O111: b4LPS (3. mu.g in 0.1ml PBS) induced endotoxin shock. 15 minutes after LPS injection, the peptide SEQ ID1 was injected at a different site (8 mg/kg/200 μ g/mouse). The mice were monitored for 48 hours and the results were recorded.
| D-galactosamine treatment | E.coli O111:B4 LPS | Peptides or buffers | Total number of mice | Survival after endotoxic shock |
| 0 | 3μg | PBS | 5 | 5(100%) |
| 20mg | 3μg | PBS | 12 | 0(0%) |
| 20mg | 3μg | SEQ ID 1 | 12 | 5(50%) |
[0087] Table 11: protection against fatal endotoxemia in semi-lactosamine-sensitized CD-1 mice is achieved by cationic peptides. CD-1 mice (9 weeks old) were made allergic to endotoxin by intraperitoneal injection of galactosamine (20mg, dissolved in 0.1ml sterile PBS). Coli O111: b4LPS (2. mu.g in 0.1ml PBS) induced endotoxin shock. 15 minutes after LPS injection, the peptide was injected at a different site (8 mg/kg at 200 μ g/mouse). The mice were monitored for 48 hours and the results were recorded.
| Peptide treatment | Coli o111 added: b4LPS | Number of mice | Survival (%) |
| Control (no peptide) | 2μg | 5 | 0 |
| SEQ ID 6 | 2μg | 5 | 40 |
| SEQ ID 13 | 2μg | 5 | 20 |
| SEQ ID 17 | 2μg | 5 | 40 |
| SEQ ID 24 | 2μg | 5 | 0 |
| SEQ ID 27 | 2μg | 5 | 20 |
[0088] Table 12: protection against fatal endotoxemia in the galena-sensitized BALB/c mice is achieved by cationic peptides. BALB/c mice (8 weeks old) were made allergic to endotoxin by intraperitoneal injection of galactosamine (20mg, dissolved in 0.1ml sterile PBS). Coli O111: b4LPS (2. mu.g in 0.1ml PBS) induced endotoxin shock. 15 minutes after LPS injection, the peptide was injected at a different site (8 mg/kg at 200 μ g/mouse). The mice were monitored for 48 hours and the results were recorded.
| Peptide treatment | Coli o111 added: b4LPS | Number of mice | Survival (%) |
| Without peptides | 2μg | 10 | 10 |
| SEQ ID 1 | 2μg | 6 | 17 |
| SEQ ID 3 | 2μg | 6 | 0 |
| SEQ ID 5 | 2μg | 6 | 17 |
| SEQ ID 6 | 2μg | 6 | 17 |
| SEQ ID 12 | 2μg | 6 | 17 |
| SEQ ID 13 | 2μg | 6 | 33 |
| SEQ ID 15 | 2μg | 6 | 0 |
| SEQ ID 16 | 2μg | 6 | 0 |
| SEQ ID 17 | 2μg | 6 | 17 |
| SEQ ID 23 | 2μg | 6 | 0 |
| SEQ ID 24 | 2μg | 6 | 17 |
| SEQ ID 26 | 2μg | 6 | 0 |
| SEQ ID 27 | 2μg | 6 | 50 |
| SEQ ID 29 | 2μg | 6 | 0 |
| SEQ ID 37 | 2μg | 6 | 0 |
| SEQ ID 38 | 2μg | 6 | 0 |
| SEQ ID 41 | 2μg | 6 | 0 |
| SEQ ID 44 | 2μg | 6 | 0 |
| SEQ ID 45 | 2μg | 6 | 0 |
[0089] Table 13: BALB/c mice were protected against fatal endotoxemia by SEQ ID 1. Coli O111: b4LPS was injected intraperitoneally into BALB/c mice. Peptides were injected at a different peritoneal site (200 μ g/mouse ═ 8 mg/kg). The mice were monitored for 48 hours and the results were recorded.
| Peptide treatment | Coli o111 added: b4LPS | Number of mice | Survival (%) |
| Without peptides | 400μg | 5 | 0 |
| SEQ ID 1 | 400μg | 5 | 100 |
[0090] Table 14: the peptides inhibit TNF- α production induced by LTA of Staphylococcus aureus. RAW264.7 murine macrophages were stimulated with 1 μ g/ml staphylococcus aureus LPS in the presence and absence of increasing concentrations of peptide. Supernatants were collected and TNF-. alpha.levels were determined by ELISA. Background levels of TNF- α production from RAW264.7 cells cultured for 6 hours without stimulation were 0.037-0.192 ng/ml. Data are presented as mean + standard deviation of three or more experiments.
| Added SEQ ID 1(μ g/ml) | Inhibition of TNF-alpha (%) |
| 0.1 | 44.5±12.5 |
| 1 | 76.7±6.4 |
| 5 | 91±1 |
| 10 | 94.5±1.5 |
| 20 | 96±1 |
[0091] Table 15: the peptide inhibits TNF-alpha production induced by non-capped lipid arabinomannan of mycobacteria. RAW264.7 murine macrophages were stimulated with 1 μ g/ml AraLAM in the presence of 20 μ g/ml peptide or polymyxin b (polymyxin b) or without peptide. Supernatants were collected and TNF-. alpha.levels were determined by ELISA. Background levels of TNF- α production from RAW264.7 cells cultured for 6 hours without stimulation were 0.037-0.192 ng/ml. Data are presented as mean + standard deviation of three or more experiments.
| Peptide (20. mu.g/ml) | Inhibition of TNF-alpha (%) |
| Without peptides | 0 |
| SEQ ID 1 | 64±5.9 |
| Polymyxin B | 15±2 |
Example 3
Evaluation of toxicity of cationic peptides
[0092]Two methods were used to measure the potential toxicity of the peptides. First, the assay was performed using a Cytotoxicity Detection Kit (Roche) (lactate dehydrogenase-LDH). This is a colorimetric assay for the quantification of cell death and cell lysis based on the measurement of LDH activity released from the cytosol of damaged cells into the supernatant. LDH is a stable cytoplasmic enzyme present in all cells that is released into the supernatant of cell cultures when the plasma membrane is damaged. An increase in the number of dead or plasma membrane-damaged cells leads to an increase in LDH enzyme activity in the culture supernatant, and OD can be measured using an ELISA plate recorder490nm(the amount of color formed in this assay is directly proportional to the number of lysed cells). In this assay, human bronchial epithelial cells (16HBEo14, HBE) were incubated with 100 μ g of peptide for 24 hours, the supernatant removed and assayed for LDH. Another assay used to measure toxicity of cationic peptides is the WST-1 assay (Roche). The assay is a colorimetric assay for the quantification of cell proliferation and cell viability based on the ability of mitochondrial dehydrogenase in living cells to cleave tetrazolium salt WST-1 (as "para")3H]-substitution of thymidine uptake assay, which is not radioactive). In this assay, HBE cells were incubated with 100. mu.g of peptide for 24 hours and then 10. mu.l was added per wellCell Proliferation Reagent (Cell Proliferation Reagent) WST-1. Cells were incubated with reagents and OD was measured using an ELISA plate recorder490nm。
[0093] The results shown below in tables 16 and 17 indicate that most of the peptides are not toxic to the cells tested. However, the results of the two analyses indicated that four of the peptides of formula F (SEQ ID NOS: 40, 41, 42 and 43) indeed caused membrane damage.
[0094] Table 16: toxicity of the cationic peptides was measured by LDH release assay. Human HBE bronchial epithelial cells were incubated with 100. mu.g/ml peptide or polymyxin B for 24 hours. LDH activity was measured in cell culture supernatants. As a control, Triton X-100 was added to release 100% LDH. Data are presented as mean ± standard deviation. Only peptides SEQ ID40, 41, 42 and 43 showed some significant toxicity.
| Treatment of | LDH Release (OD)490nm) |
| Cell-free controls | 0.6±0.1 |
| Control of Triton X-100 | 4.6±0.1 |
| Control without peptide | 1.0±0.05 |
| SEQ ID 1 | 1.18±0.05 |
| SEQ ID 3 | 1.05±0.04 |
| SEQ ID 6 | 0.97±0.02 |
| SEQ ID 7 | 1.01±0.04 |
| SEQ ID 9 | 1.6±0.03 |
| SEQ ID 10 | 1.04±0.04 |
| SEQ ID 13 | 0.93±0.06 |
| SEQ ID 14 | 0.99±0.05 |
| SEQ ID 16 | 0.91±0.04 |
| SEQ ID 17 | 0.94±0.04 |
| SEQ ID 19 | 1.08±0.02 |
| SEQ ID 20 | 1.05±0.03 |
| SEQ ID 21 | 1.06±0.04 |
| SEQ ID 22 | 1.29±0.12 |
| SEQ ID 23 | 1.26±0.46 |
| SEQ ID 24 | 1.05±0.01 |
| SEQ ID 26 | 0.93±0.04 |
| SEQ ID 27 | 0.91±0.04 |
| SEQ ID 28 | 0.96±0.06 |
| SEQ ID 29 | 0.99±0.02 |
| SEQ ID 31 | 0.98±0.03 |
| SEQ ID 33 | 1.03±0.05 |
| SEQ ID 34 | 1.02±0.03 |
| SEQ ID 35 | 0.88±0.03 |
| SEQ ID 36 | 0.85±0.04 |
| SEQ ID 37 | 0.96±0.04 |
| SEQ ID 38 | 0.95±0.02 |
| SEQ ID 40 | 2.8±0.5 |
| SEQ ID 41 | 3.3±0.2 |
| SEQ ID 42 | 3.4±0.2 |
| SEQ ID 43 | 4.3±0.2 |
| SEQ ID 44 | 0.97±0.03 |
| SEQ ID 45 | 0.98±0.04 |
| SEQ ID 47 | 1.05±0.05 |
| SEQ ID 48 | 0.95±0.05 |
| SEQ ID 53 | 1.03±0.06 |
| Polymyxin B | 1.21±0.03 |
[0095] Table 17: toxicity of the cationic peptides was measured by WST-1 analysis. HBE cells were incubated with 100. mu.g/ml peptide or polymyxin B for 24 hours and cell survival was then measured. Data are presented as mean ± standard deviation. As a control, Triton X-100 was added to release 100% LDH. Only peptides SEQ ID40, 41, 42 and 43 showed some significant toxicity.
| Treatment of | OD490nm |
| Cell-free controls | 0.24±0.01 |
| Control of Triton X-100 | 0.26±0.01 |
| Control without peptide | 1.63±0.16 |
| SEQ ID 1 | 1.62±0.34 |
| SEQ ID 3 | 1.35±0.12 |
| SEQ ID 10 | 1.22±0.05 |
| SEQ ID 6 | 1.81±0.05 |
| SEQ ID 7 | 1.78±0.10 |
| SEQ ID 9 | 1.69±0.29 |
| SEQ ID 13 | 1.23±0.11 |
| SEQ ID 14 | 1.25±0.02 |
| SEQ ID 16 | 1.39±0.26 |
| SEQ ID 17 | 1.60±0.46 |
| SEQ ID 19 | 1.42±0.15 |
| SEQ ID 20 | 1.61±0.21 |
| SEQ ID 21 | 1.28±0.07 |
| SEQ ID 22 | 1.33±0.07 |
| SEQ ID 23 | 1.14±0.24 |
| SEQ ID 24 | 1.27±0.16 |
| SEQ ID 26 | 1.42±0.11 |
| SEQ ID 27 | 1.63±0.03 |
| SEQ ID 28 | 1.69±0.03 |
| SEQ ID 29 | 1.75±0.09 |
| SEQ ID 31 | 1.84±0.06 |
| SEQ ID 33 | 1.75±0.21 |
| SEQ ID 34 | 0.96±0.05 |
| SEQ ID 35 | 1.00±0.08 |
| SEQ ID 36 | 1.58±0.05 |
| SEQ ID 37 | 1.67±0.02 |
| SEQ ID 38 | 1.83±0.03 |
| SEQ ID 40 | 0.46±0.06 |
| SEQ ID 41 | 0.40±0.01 |
| SEQ ID 42 | 0.39±0.08 |
| SEQ ID 43 | 0.46±0.10 |
| SEQ ID 44 | 1.49±0.39 |
| SEQ ID 45 | 1.54±0.35 |
| SEQ ID 47 | 1.14±0.23 |
| SEQ ID 48 | 0.93±0.08 |
| SEQ ID 53 | 1.51±0.37 |
| Polymyxin B | 1.30±0.13 |
Example 4
Polynucleotide regulation by cationic peptides
[0096]The effect of the cationic peptide itself on the transcriptional response of macrophages and epithelial cells was measured using a polynucleotide array. Mouse macrophage RAW264.7, human bronchial cell (HBE) or A549 human epithelial cellCells were plated on 150mm tissue culture dishes at a density of 5.6X 10 cells per dish6Cells were cultured overnight and then incubated with 50. mu.g/ml peptide for 4 hours or only with medium without peptide. After stimulation, cells were washed once with diethylpyrocarbonate-treated PBS and scraped from the culture dish with a cell brush. Total RNA was isolated using Trizol (Gibco Life technologies). The RNA pellet was resuspended in RNase-free water containing RNase inhibitor (Ambion, Austin, TX). The RNA was treated with DNaseI (Clontech, Palo alto, Calif.) at 37 ℃ for one hour. Add stop mixture (0.1M EDTA [ pH 8.0 ]]1mg/ml glycogen), the sample was washed once with phenol: chloroform: isoamyl alcohol (25: 24: 1) and once with chloroform. Then 2.5 volumes of 100% ethanol and 1/10 volumes of sodium acetate were added and the RNA was precipitated at pH 5.2. The RNA was resuspended in RNase-free water containing RNase inhibitor (Ambion) and stored at-70 ℃. The quality of the RNA was estimated by electrophoresis on a 1% agarose gel. Using the isolated RNA as a template, PCR amplification was performed using β actin-specific primers 5'-GTCCCTGTATGCCTCTGGTC-3' (SEQ ID NO: 55) and 5'-GATGTCACGCACGATTTCC-3' (SEQ ID NO: 56)) to determine whether genomic DNA was contaminated. Agarose gel electrophoresis and ethidium bromide staining confirmed that no amplification product was present after 35 cycles.
[0097]Atlas cDNA expression array (Clontech, Palo Alto, Calif.) consisted of 588 via-selected murine cDNAs spotted in duplicate (duplcate) on a positively charged membrane, which were used in earlier polynucleotide array studies (Table 18, 19)。Prepared from 5 mu g of total RNA32p radiolabeled cDNA probes, incubated with the array at 71 ℃ overnight. A flood wash was performed and then exposed to a phosphoimager system (molecular dynamics, Sunnyvale, Calif.) for 3 days at 4 ℃. Images were acquired using Molecular Dynamics PSI phosphor screen imaging (phosphor). Hybridization signals were analyzed using atlas image 1.0 image analysis software (Clontech) and Excel (Microsoft, Redmond, WA). The intensity of each spot was corrected for background levels and normalized for differences between probe labels by using the intensity found in the stimulusAverage of 5 polynucleotides with little change between conditions: beta actin, ubiquitin, ribosomal protein S29, glyceraldehyde-3-phosphate dehydrogenase (GAPDH) and Ca2+Binding proteins. When the normalized hybridization signal for a given cDNA is less than 20, it is assigned a value of 20, from which the ratio and relative expression are calculated.
[0098] The polynucleotide array used in the next step (tables 21-26) is a Resgen Human cDNA array (identification number of the genome is PRHU03-S3) which consists of duplicate 7,458 personal cDNA spots. Probes were prepared with 15-20. mu.g total RNA and labeled with Cy 3-labeled dUTP. The probe was purified and hybridized to a printed glass slide, overnight at 42 ℃ and then washed. After washing, images were taken with a Virtek slide recorder. The mean, median and background intensities of the spots were determined using image processing software (Imagene 4.1, Marina Del Rey, Calif.). The normalization process and analysis were performed using Genespring software (Redwood City, Calif.). The average background intensity was subtracted from the average intensity measured by Imagene to calculate the intensity value. The intensity of each spot was normalized by obtaining the median spot intensity from a set of values for the spot within one slide and comparing that value to the values for all slides in this experiment. The relative change between the peptide-treated cells and the control cells can be seen in the table below.
[0099]Other polynucleotide arrays used (tables 27-35) were the Human Operon array (identification number of the genome is PRHU04-S1), which consists of duplicate dots of 14,000 individual oligomers. Probes were prepared with 10. mu.g total RNA and labeled with Cy3 or Cy5 labeled dUTP. In these experiments, A549 epithelial cells were plated into 100mm tissue culture dishes at a density of 2.5X 10 per dish6And (4) cells. Total RNA was isolated using RNAqueous (Ambion). DNA contamination was removed using a DNA removal kit (Ambion). Probes prepared from total RNA were purified and hybridized to printed glass slides, overnight at 42 ℃ and then washed. After washing, images were taken with a Perkin Elmer array scanner. The mean intensity of the spots was determined by image processing software (Imagene 5.0, Marina Del Rey, Calif.)Degree, intermediate intensity and background intensity. The background was removed using a "homemade" procedure. The program calculates the base intensity of each sub-cell as 10% and subtracts this value for each cell. The analysis was performed using Genespring software (Redwood City, Calif.). The intensity of each spot was normalized by obtaining the median spot intensity from a set of values for the spot within one slide and comparing that value to the values for all slides in this experiment. The relative change between the peptide-treated cells and the control cells can be seen in the table below.
[00100]Semi-quantitative RT-PCR was performed to confirm the results of the polynucleotide array. Mu.g of RNA sample and 1. mu.l of oligo dT (500. mu.g/ml) and 1. mu.l of mixed dNTP stock at a concentration of 1mM were incubated at 65 ℃ for 5 minutes in a reaction volume of 12. mu.l using water treated with Diethylpyrocarbonate (DEPC) in a thermal cycler. Mu.15 XFirst-Strand buffer, 2. mu.l of 0.1M DTT and 1. mu.l RNaseOUT recombinant ribonuclease inhibitor (40 units/. mu.l) were added, incubated at 42 ℃ for 2 minutes, and then 1. mu.l (200units) of Superscript II (Invitrogen, Burlington, ON) was added. For each cDNA source, a parallel experiment was performed without Superscript II to provide a negative control. Using 5 'and 3' primers (1.0. mu.M), 0.2mM dNTP mix, 1.5mM MgCl2cDNA was amplified with 1U Taq DNA polymerase (New England Biolabs, Missiauga, ON) and 1 XPCR buffer. Each PCR was performed on a thermal cycler, comprising 30-40 cycles, each cycle comprising denaturation at 94 ℃ for 30 seconds, annealing at 52 ℃ or 55 ℃ for 30 seconds, and extension at 72 ℃ for 40 seconds. The number of cycles of PCR was optimized for each primer and each RNA sample to lie within the linear range of the reaction. To evaluate the extraction step and estimate the amount of RNA, the housekeeping (housekeeping) polynucleotide β actin gene was amplified in each experiment. The reaction products were visualized by electrophoresis and analyzed by densitometry (densitometry), which allows the relative concentration of the starting RNA to be calculated with reference to the amplification of the beta actin polynucleotide.
[00101] Table 18 shows that, in the selected known polynucleotides, on the small Atlas microarray, the sequence of SEQ ID NO: 1 treatment of RAW264.7 cells upregulated the expression of more than 30 different polynucleotides therein. The peptide consisting of SEQ ID NO: 1 the upregulated polynucleotides come primarily from two classes: one class includes receptors (growth factors, chemokines, interleukins, interferons, hormones, neurotransmitters), cell surface antigens and cell adhesion, and the other class includes cell-cell communication (growth factors, cytokines, chemokines, interleukins, interferons, hormones), cytoskeleton, cell motility and protein turnover. Specific polynucleotides that are upregulated include polynucleotides encoding the following proteins: chemokines MCP-3, anti-inflammatory cytokines IL-10, macrophage colony stimulating factors and receptors such as IL-1R-2 (a recognized antagonist of prolific IL-1 that binds to IL-1R 1), PDGF receptor B, NOTCH4, LIF receptor, LFA-1, TGF beta receptor 1, G-CSF receptor, and IFN gamma receptor. The peptides also up-regulate polynucleotides encoding several metalloproteinases and their inhibitors, including bone morphogenetic proteins BMP-1, BMP-2, BMP-8a, TIMP 2 and TIMP 3. Moreover, the peptide upregulates several specific transcription factors, including JunD, as well as YY and LIM-1 transcription factors, and kinases such as Etk1 and Csk, suggesting that it has a broad effect. The study of polynucleotide arrays also found that seq id NO: 1 down-regulated at least 20 polynucleotides in RAW264.7 macrophages (table 19). Polynucleotides downregulated by peptides include DNA repair proteins and several inflammatory mediators such as MIP-1 α, oncostatin M, and IL-12. Many of the effects of the peptides on polynucleotide expression were confirmed by RT-PCR (Table 20). Using a medium-sized microarray (7835 polynucleotides), it was also found that the peptide SEQ ID NO: 2. SEQ ID NO: 3. SEQ ID NO: 19 and SEQ ID NO: 1, and representative peptides of each formula, alter transcriptional responses in human epithelial cell lines. SEQ ID NO: 1 effects on polynucleotide expression were compared in two human epithelial cell lines, A549 and HBE. Table 21 describes polynucleotides related to host immune responses that are up-regulated by 2 or more peptides at a 2-fold greater rate compared to unstimulated cells. Table 22 describes polynucleotides associated with host immune responses that are downregulated by 2 or more peptides at a rate of more than 2-fold compared to unstimulated cells. Tables 23 and 24 show up-regulated and down-regulated human epithelial pro-inflammatory polynucleotides, respectively. Tables 25 and 26 show the pro-inflammatory polynucleotides affected by the cationic peptide. One clear trend is that cationic peptides up-regulate the anti-inflammatory response, down-regulate the pro-inflammatory response. It was very difficult to find a polynucleotide that was associated with an anti-inflammatory response, but was down-regulated (Table 26). Proinflammatory polynucleotides that are upregulated by cationic peptides are primarily polynucleotides associated with migration and adhesion. It should be noted that among the proinflammatory polynucleotides that are down-regulated, there are several toll-like receptor (TLR) polynucleotides affected by all cationic peptides, which TLR polynucleotides are important for signaling a host's response to an infectious agent. An important anti-inflammatory polynucleotide that is upregulated by all peptides is the IL-10 receptor polynucleotide. IL-10 is an important cytokine involved in the regulation of proinflammatory cytokines. For the peptide SEQ ID NO: these effects on polynucleotide expression can also be observed when primary human macrophages are used, as shown in tables 27 and 28. Tables 31-37 below show the effect of representative peptides of each formula on the expression of selected polynucleotides (14,000 tested) by human epithelial cells. To test the ability of the peptides to alter the expression of the human epithelial polynucleotide, at least 6 peptides were selected for each formula, which did have a broad stimulatory effect. For each formula, typically at least 50 polynucleotides are up-regulated by each peptide in the set.
[00102]Table 18: peptide SEQ ID NO: 1 treatment of the Up-regulated polynucleotidea. Cationic peptides at a concentration of 50. mu.g/ml were found to efficiently induce the expression of several polynucleotides. Peptides were incubated with RAW cells for 4 hours, RNA was isolated, converted to labeled cDNA probes, and hybridized to Atlas arrays. The intensity of the unstimulated cells is shown in the third column. "ratio peptide: by unstimulated "column is meant the intensity of polynucleotide expression in cells stimulated with the peptide divided by the intensity of the unstimulated cells.
[00103] The normalized intensities of housekeeping polynucleotides vary from 0.8 to 1.2 fold, and thus these polynucleotides can be effectively used in the normalization process. When the standard hybridization intensity of a given cDNA is below 20, the value is assigned to 20, from which the ratio and relative expression are calculated. The assay was repeated three times with different RNA preparations, with the average fold change shown below. Polynucleotides that vary by a factor of two or more relative to expression levels are shown.
| Polynucleotides/proteins | Polynucleotide function | Intensity of non-stimulation | Ratio peptide: is not stimulated | Sequence accession number |
| Etk 1 | Tyrosine protein kinase receptors | 20 | 43 | M68513 |
| PDGFRB | Growth factor receptors | 24 | 25 | X04367 |
| Corticotropin releasing factor receptors | 20 | 23 | X72305 | |
| NOTCH4 | Proto-cancer polynucleotide | 48 | 18 | M80456 |
| IL-1R2 | Interleukin receptors | 20 | 16 | X59769 |
| MCP-3 | Chemotactic factor | 56 | 14 | S71251 |
| BMP-1 | Bone morphogenetic proteins | 20 | 14 | L24755 |
| Endothelin b receptor | Receptors | 20 | 14 | U32329 |
| c-ret | Cancer polynucleotide precursors | 20 | 13 | X67812 |
| LIFR | Cytokine receptors | 20 | 12 | D26177 |
| BMP-8a | Bone morphogenetic proteins | 20 | 12 | M97017 |
| Zfp92 | Zinc finger protein 92 | 87 | 11 | U47104 |
| MCSF | Macrophage colony stimulating factor 1 | 85 | 11 | X05010 |
| GCSFR | Granulocyte colony stimulating factor receptor | 20 | 11 | M58288 |
| IL-8RB | Chemokine receptors | 112 | 10 | D17630 |
| IL-9R | Interleukin receptors | 112 | 6 | M84746 |
| Cas | Crk-related substrates | 31 | 6 | U48853 |
| p58/GTA | Kinase enzymes | 254 | 5 | M58633 |
| CASP2 | Caspase (caspase) precursors | 129 | 5 | D28492 |
| IL-1 beta precursor | Interleukin precursors | 91 | 5 | M15131 |
| SPI2-2 | Serine protease inhibitors | 62 | 5 | M64086 |
| C5AR | Chemokine receptors | 300 | 4 | S46665 |
| L-myc | Cancer polynucleotides | 208 | 4 | X13945 |
| IL-10 | Interleukin | 168 | 4 | M37897 |
| p19ink4 | cdk4 and cdk6 inhibitors | 147 | 4 | U19597 |
| ATOH2 | Non-regulated Gene homolog 2 | 113 | 4 | U29086 |
| DNAsel | DNA enzyme | 87 | 4 | U00478 |
| CXCR-4 | Chemokine receptors | 36 | 4 | D87747 |
| Cyclin D3 | Cyclin proteins | 327 | 3 | U43844 |
| IL-7Rα | Interleukin receptors | 317 | 3 | M29697 |
| POLA | DNA polymerase alpha | 241 | 3 | D17384 |
| Tie-2 | Cancer polynucleotides | 193 | 3 | S67051 |
| DNL1 | DNA ligase I | 140 | 3 | U04674 |
| BAD | Apoptosis proteins | 122 | 3 | L37296 |
| GADD45 | Protein capable of inducing DNA damage | 88 | 3 | L28177 |
| Sik | Scr-related kinase | 82 | 3 | U16805 |
| Integrin 4 | Integrin | 2324 | 2 | X53176 |
| TGFβR1 | Growth factor receptors | 1038 | 2 | D25540 |
| LAMR1 | Receptors | 1001 | 2 | J02870 |
| Crk | Crk adaptor protein | 853 | 2 | S72408 |
| ZFX | Chromosomal proteins | 679 | 2 | M32309 |
| Cyclin E1 | Cyclin proteins | 671 | 2 | X75888 |
| POLD1 | Subunit of DNA polymerase | 649 | 2 | Z21848 |
| Vav | Proto-cancer polynucleotide | 613 | 2 | X64361 |
| YY(NF-E1) | Transcription factor | 593 | 2 | L13968 |
| JunD | Transcription factor | 534 | 2 | J050205 |
| Csk | c-src kinase | 489 | 2 | U05247 |
| Cdk7 | Cyclin dependent kinases | 475 | 2 | U11822 |
| MLC1A | Myosin light subunit isoforms | 453 | 2 | M19436 |
| ERBB-3 | Receptors | 435 | 2 | L47240 |
| UBF | Transcription factor | 405 | 2 | X60831 |
| TRAIL | Apoptosis ligands | 364 | 2 | U37522 |
| LFA-1 | Cell adhesion receptors | 340 | 2 | X14951 |
| SLAP | Src-like adaptor proteins | 315 | 2 | U29056 |
| IFNGR | Interferon gamma receptors | 308 | 2 | M28233 |
| LIM-1 | Transcription factor | 295 | 2 | Z27410 |
| ATF-2 | Transcription factor | 287 | 2 | S76657 |
| FST | Follistatin precursors | 275 | 2 | Z29532 |
| TIMP3 | Protease inhibitors | 259 | 2 | L19622 |
| RU49 | Transcription factor | 253 | 2 | U41671 |
| IGF-1Rα | Insulin-like growth factor receptors | 218 | 2 | U00182 |
| Cyclin G2 | Cyclin proteins | 214 | 2 | U95826 |
| fyn | Tyrosine protein kinase | 191 | 2 | U70324 |
| BMP-2 | Bone morphologyProduced protein | 186 | 2 | L25602 |
| Brn-3.2 POU | Transcription factor | 174 | 2 | S68377 |
| KIF1A | Kinesin family proteins | 169 | 2 | D29951 |
| MRC1 | Mannose receptor | 167 | 2 | Z11974 |
| PAI2 | Protease inhibitors | 154 | 2 | X19622 |
| BKLF | CACCC sequence frame binding protein | 138 | 2 | U36340 |
| TIMP2 | Protease inhibitors | 136 | 2 | X62622 |
| Mas | Protooncogene | 131 | 2 | X67735 |
| NURR-1 | Transcription factor | 129 | 2 | S53744 |
[00104]Table 19: the expression of the polypeptide encoded by SEQ ID NO: 1 treatment of Down-regulated polynucleotidesa. Cationic peptides at a concentration of 50. mu.g/ml were found to reduce the expression of several polynucleotides. Peptides were incubated with RAW cells for 4 hours, RNA was isolated, converted to labeled cDNA probes, and hybridized to Atlas arrays. The intensity of the unstimulated cells is shown in the third column. "ratio peptide: by unstimulated "column is meant the intensity of polynucleotide expression in cells stimulated with the peptide divided by the intensity of the unstimulated cells. The assay was repeated three times with different cells and the mean fold change is shown below. Polynucleotides that vary approximately two-fold or more in relative expression levels are shown.
| Polynucleotides/proteins | Polynucleotide function | Intensity of non-stimulation | Ratio peptide: is not stimulated | Sequence accession number |
| Sodium channels | Voltage-gated ion channels | 257 | 0.08 | L36179 |
| XRCC1 | DNA repair proteins | 227 | 0.09 | U02887 |
| est-2 | Cancer polynucleotides | 189 | 0.11 | J04103 |
| XPAC | DNA repair proteins | 485 | 0.12 | X74351 |
| EPOR | Receptor precursors | 160 | 0.13 | J04843 |
| PEA 3 | Ets related proteins | 158 | 0.13 | X63190 |
| Orphan receptors | Nuclear receptors | 224 | 0.2 | U11688 |
| N-cadherins | Cell adhesion receptors | 238 | 0.23 | M31131 |
| OCT3 | Transcription factor | 583 | 0.24 | M34381 |
| PLCβ | Phospholipase enzymes | 194 | 0.26 | U43144 |
| KRT18 | Intermediate fibrillar proteins | 318 | 0.28 | M11686 |
| THAM | Enzyme | 342 | 0.32 | X58384 |
| CD40L | CD40 ligand | 66 | 0.32 | X65453 |
| CD86 | T lymphocyte antigens | 195 | 0.36 | L25606 |
| Oncostatin M | Cytokine | 1127 | 0.39 | D31942 |
| PMS2 DNA | DNA repair proteins | 200 | 0.4 | U28724 |
| IGFBP6 | Growth factor | 1291 | 0.41 | X81584 |
| MIP-1β | Cytokine | 327 | 0.42 | M23503 |
| ATBF1 | AT motif binding factor | 83 | 0.43 | D26046 |
| Nucleobindin (nuclear binding protein) | Golgi resident proteins | 367 | 0.43 | M96823 |
| bcl-x | Apoptosis proteins | 142 | 0.43 | L35049 |
| Uromodulin (uromollin) | Glycoprotein | 363 | 0.47 | L33406 |
| IL-12 p40 | Interleukin | 601 | 0.48 | M86671 |
| MmRad52 | DNA repair proteins | 371 | 0.54 | Z32767 |
| Tob1 | Anti-proliferative factor | 956 | 0.5 | D78382 |
| Ung1 | DNA repair proteins | 535 | 0.51 | X99018 |
| KRT19 | Intermediate fibrillar proteins | 622 | 0.52 | M28698 |
| PLCγ | Phospholipase enzymes | 251 | 0.52 | X95346 |
| Integrin alpha6 | Cell adhesion receptors | 287 | 0.54 | X69902 |
| GLUT1 | Glucose transporters | 524 | 0.56 | M23384 |
| CTLA4 | Immunoglobulin superfamily | 468 | 0.57 | X05719 |
| FRA2 | Fos-associated antigens | 446 | 0.57 | X83971 |
| MTRP | Lysosomal associated proteins | 498 | 0.58 | U34259 |
[00105] Table 20: in the case of the peptide SEQ ID NO: 1 can be confirmed by RT-PCR. RAW264.7 macrophages were incubated with 50. mu.g/ml peptide for 4 hours or separately with culture medium for 4 hours. Total RNA was isolated and subjected to semi-quantitative RT-PCR. Primer pairs specific for each polynucleotide were used to amplify RNA. Amplification of beta actin was used as a positive control and was used for normalization. Density measurement analysis was performed on the RT-PCR products. The results are expressed as the relative fold change in polynucleotide expression for the peptide-treated cells compared to cells incubated with medium alone. Data are presented as mean ± standard deviation of three experiments.
| Polynucleotide | Array ratio-* | RT-PCR-* |
| CXCR-4 | 4.0±1.7 | 4.1±0.9 |
| IL-8RB | 9.5±7.6 | 7.1±1.4 |
| MCP-3 | 13.5±4.4 | 4.8±0.88 |
| IL-10 | 4.2±2.1 | 16.6±6.1 |
| CD14 | 0.9±0.1 | 0.8±0.3 |
| MIP-1B | 0.42±0.09 | 0.11±0.04 |
| XRCC1 | 0.12±0.01 | 0.25±0.093 |
| MCP-1 | Without arrays | 3.5±1.4 |
[00106]Table 21: polynucleotides upregulated by peptide treatment in A549 epithelial cellsa. Cationic peptides at a concentration of 50. mu.g/ml were found to increase the expression of several polynucleotides. The peptides were incubated with human A549 epithelial cells for 4 hours, RNA was isolated, converted to labeled cDNA probes, and hybridized to human cDNA array ID # PRHU 03-S3. The intensity of the polynucleotide in the unstimulated cells is shown in the second column. "ratio peptide: by unstimulated "column is meant the intensity of polynucleotide expression in cells stimulated with the peptide divided by the intensity of the unstimulated cells.
| Polynucleotides/proteins | Strength of not being stimulated | Ratio peptide: is not stimulated | Registration number | |||
| ID 2 | ID 3 | ID 19 | ID 1 | |||
| IL-1R antagonist homolog 1 | 0.00 | 3086 | 1856 | 870 | AI167887 | |
| IL-10Rβ | 0.53 | 2.5 | 1.6 | 1.9 | 3.1 | AA486393 |
| IL-11Rα | 0.55 | 2.4 | 1.0 | 4.9 | 1.8 | AA454657 |
| IL-17R | 0.54 | 2.1 | 2.0 | 1.5 | 1.9 | AW029299 |
| TNF R superfamily, member 1B | 0.28 | 18 | 3.0 | 15 | 3.6 | AA150416 |
| TNF R superfamily, member 5(CD40LR) | 33.71 | 3.0 | 0.02 | H98636 | ||
| TNF R superfamily, member 11b | 1.00 | 5.3 | 4.50 | 0.8 | AA194983 | |
| IL-8 | 0.55 | 3.6 | 17 | 1.8 | 1.1 | AA102526 |
| Interleukin enhancer binding factor 2 | 0.75 | 1.3 | 2.3 | 0.8 | 4.6 | AA894687 |
| Interleukin enhancer binding factor 1 | 0.41 | 2.7 | 5.3 | 2.5 | R56553 | |
| Protein containing SH2 and capable of being induced by cell factor | 0.03 | 33 | 44 | 39 | 46 | AA427521 |
| IK cytokines, down-regulators of HLA II | 0.50 | 3.1 | 2.0 | 1.7 | 3.3 | R39227 |
| Protein containing SH2 and capable of being induced by cell factor | 0.03 | 33 | 44 | 39 | 46 | AA427521 |
| Small molecule inducible cytokine subfamily A (Cys-Cys), member 21 | 1.00 | 3.9 | 2.4 | AI922341 | ||
| TGFB-induced early growth response 2 | 0.90 | 2.4 | 2.1 | 0.9 | 1.1 | AI473938 |
| NK cell R | 1.02 | 2.5 | 0.7 | 0.3 | 1.0 | AA463248 |
| CCR6 | 0.14 | 4.5 | 7.8 | 6.9 | 7.8 | N57964 |
| Cell adhesion molecules | 0.25 | 4.0 | 3.9 | 3.9 | 5.1 | R40400 |
| Melanoma adhesion molecules | 0.05 | 7.9 | 20 | 43 | 29.1 | AA497002 |
| CD31 | 0.59 | 2.7 | 3.1 | 1.0 | 1.7 | R22412 |
| Integrin,. alpha.2 (CD49B,. alpha.2 subunit of the VLA-2 receptor) | 1.00 | 0.9 | 2.4 | 3.6 | 0.9 | AA463257 |
| Integrin,. alpha.3 (CD49C, VLA-3) | 0.94 | 0.8 | 2.5 | 1.9 | 1.1 | AA424695 |
| The alpha 3 subunit of the receptor) | ||||||
| Integrin, alpha E | 0.01 | 180 | 120 | 28 | 81 | AA425451 |
| Integrin,. beta.1 | 0.47 | 2.1 | 2.1 | 7.0 | 2.6 | W67174 |
| Integrin,. beta.3 | 0.55 | 2.7 | 2.8 | 1.8 | 1.0 | AA037229 |
| Integrin,. beta.3 | 0.57 | 2.6 | 1.4 | 1.8 | 2.0 | AA666269 |
| Integrin,. beta.4 | 0.65 | 0.8 | 2.2 | 4.9 | 1.5 | AA485668 |
| Integrin beta 4 binding proteins | 0.20 | 1.7 | 5.0 | 6.6 | 5.3 | AI017019 |
| Calcium and integrin binding proteins | 0.21 | 2.8 | 4.7 | 9.7 | 6.7 | AA487575 |
| Disintegrin and metalloprotease domain 8 | 0.46 | 3.1 | 2.2 | 3.8 | AA279188 | |
| Disintegrin and metalloprotease domain 9 | 0.94 | 1.1 | 2.3 | 3.6 | 0.5 | H59231 |
| Disintegrin and metalloprotease domain 10 | 0.49 | 1.5 | 2.1 | 3.3 | 2.2 | AA043347 |
| Disintegrin and metalloprotease domain 23 | 0.44 | 1.9 | 2.3 | 2.5 | 4.6 | H11006 |
| Cadherin type 1, E-cadherin (epithelium) | 0.42 | 8.1 | 2.2 | 2.4 | 7.3 | H97778 |
| Cadherin type 12, 2 (N-cadherin 2) | 0.11 | 13 | 26 | 9.5 | AI740827 | |
| Procadherin 12 | 0.09 | 14.8 | 11.5 | 2.6 | 12.4 | AI652584 |
| Procadherin gamma subfamily C, 3 | 0.34 | 3.0 | 2.5 | 4.5 | 9.9 | R89615 |
| Catenin (cadherin-related protein), delta 1 | 0.86 | 1.2 | 2.2 | 2.4 | AA025276 | |
| Laminin R1(67kD, ribosomal protein SA) | 0.50 | 0.4 | 2.0 | 4.4 | 3.0 | AA629897 |
| Killer cell lectin-like receptor subfamily C, member 2 | 0.11 | 9.7 | 9.0 | 4.1 | 13.4 | AA190627 |
| Killer cell lectin-like receptor subfamily C, member 3 | 1.00 | 3.2 | 1.0 | 0.9 | 1.3 | W93370 |
| Killer cell lectin-like receptor subfamily G, member 1 | 0.95 | 2.3 | 1.7 | 0.7 | 1.1 | AI433079 |
| C-type lectin-like receptors2 | 0.45 | 2.1 | 8.0 | 2.2 | 5.3 | H70491 |
| CSF 3 R | 0.40 | 1.9 | 2.5 | 3.5 | 4.0 | AA458507 |
| Macrophage stimulation of 1R | 1.00 | 1.7 | 2.3 | 0.4 | 0.7 | AA173454 |
| BMP type R IA | 0.72 | 1.9 | 2.8 | 0.3 | 1.4 | W15390 |
| Formyl peptide receptor 1 | 1.00 | 3.1 | 1.4 | 0.4 | AA425767 | |
| CD2 | 1.00 | 2.6 | 0.9 | 1.2 | 0.9 | AA927710 |
| CD36 | 0.18 | 8.2 | 5.5 | 6.2 | 2.5 | N39161 |
| Vitamin D R | 0.78 | 2.5 | 1.3 | 1.1 | 1.4 | AA485226 |
| Human protease activation of R-2 | 0.54 | 6.1 | 1.9 | 2.2 | AA454652 | |
| Prostaglandin E receptor 3(EP3 subtype) | 0.25 | 4.1 | 4.9 | 3.8 | 4.9 | AA406362 |
| PDGF R beta polypeptides | 1.03 | 2.5 | 1.0 | 0.5 | 0.8 | R56211 |
| VIP R2 | 1.00 | 3.1 | 2.0 | AI057229 | ||
| Growth factor receptor binding protein 2 | 0.51 | 2.2 | 2.0 | 2.4 | 0.3 | AA449831 |
| Murine mammary tumor virus receptor homologs | 1.00 | 6.9 | 16 | W93891 | ||
| Adenosine A2a R | 0.41 | 3.1 | 1.8 | 4.0 | 2.5 | N57553 |
| Adenosine A3R | 0.83 | 2.0 | 2.3 | 1.0 | 1.2 | AA863086 |
| T cell R delta locus | 0.77 | 2.7 | 1.3 | 1.8 | AA670107 | |
| Prostaglandin E receptor 1(EP1 subtype) | 0.65 | 7.2 | 6.0 | 1.5 | AA972293 | |
| Growth factor receptor binding protein 14 | 0.34 | 3.0 | 6.3 | 2.9 | R24266 | |
| Epstein-Barr Virus induced multimerization | 0.61 | 1.6 | 2.4 | 8.3 | AA037376 |
| Nucleotide 2 | ||||||
| Complement component receptor 2 | 0.22 | 26 | 4.5 | 2.6 | 18.1 | AA521362 |
| Endotoxin receptor type A | 0.07 | 12 | 14 | 14 | 16 | AA450009 |
| v-SNARE R | 0.56 | 11 | 12 | 1.8 | AA704511 | |
| Tyrosine kinases, non-receptor, 1 | 0.12 | 7.8 | 8.5 | 10 | 8.7 | AI936324 |
| Receptor tyrosine kinase-like orphan receptor 2 | 0.40 | 7.3 | 5.0 | 1.6 | 2.5 | N94921 |
| Protein tyrosine phosphatase, non-receptor type 3 | 1.02 | 1.0 | 13.2 | 0.5 | 0.8 | AA682684 |
| Protein tyrosine phosphatase, non-receptor type 9 | 0.28 | 2.5 | 4.0 | 0.9 | 5.3 | AA434420 |
| Protein tyrosine phosphatase, non-receptor type 11 | 0.42 | 2.9 | 2.4 | 2.2 | 3.0 | AA995560 |
| Protein tyrosine phosphatase, non-receptor type 12 | 1.00 | 2.3 | 2.2 | 0.8 | 0.5 | AA446259 |
| Protein tyrosine phosphatase, non-receptor type 13 | 0.58 | 1.7 | 2.4 | 3.6 | 1.7 | AA679180 |
| Protein tyrosine phosphatase, non-receptor type 18 | 0.52 | 3.2 | 0.9 | 1.9 | 6.5 | AI668897 |
| Protein tyrosine phosphatases, receptor types, A | 0.25 | 4.0 | 2.4 | 16.8 | 12.8 | H82419 |
| Protein tyrosine phosphatases, receptor types, J | 0.60 | 3.6 | 3.2 | 1.6 | 1.0 | AA045326 |
| Protein tyrosine phosphatases, receptor types, T | 0.73 | 1.2 | 2.8 | 3.0 | 1.4 | R52794 |
| Protein tyrosine phosphatase, receptor type, U | 0.20 | 6.1 | 1.2 | 5.6 | 5.0 | AA644448 |
| Protein tyrosine phosphatases, receptor types, C-related proteins | 1.00 | 5.1 | 2.4 | AA481547 | ||
| Phospholipase A2 receptor 1 | 0.45 | 2.8 | 2.2 | 1.9 | 2.2 | AA086038 |
| MAP kinase activated protein kinase 3 | 0.52 | 2.1 | 2.7 | 1.1 | 1.9 | W68281 |
| MAP kinase 6 | 0.10 | 18 | 9.6 | 32 | H07920 | |
| MAP kinase 5 | 1.00 | 3.0 | 5.2 | 0.8 | 0.2 | W69649 |
| MAP kinase 7 | 0.09 | 11.5 | 12 | 33 | H39192 |
| MAP kinase 12 | 0.49 | 2.1 | 1.7 | 2.2 | 2.0 | AI936909 |
| G protein coupled receptor 4 | 0.40 | 3.7 | 3.0 | 2.4 | 2.5 | AI719098 |
| G protein coupled receptor 49 | 0.05 | 19 | 19 | 27 | AA460530 | |
| G protein coupled receptor 55 | 0.08 | 19 | 15 | 12 | N58443 | |
| G protein coupled receptor 75 | 0.26 | 5.2 | 3.1 | 7.1 | 3.9 | H84878 |
| G protein coupled receptor 85 | 0.20 | 6.8 | 5.4 | 4.9 | 5.0 | N62306 |
| Regulatory protein 20 for G protein signaling | 0.02 | 48 | 137 | 82 | AI264190 | |
| Regulatory protein 6 for G protein signaling | 0.27 | 3.7 | 8.9 | 10.6 | R39932 | |
| Killer protein (inducing apoptosis) acting with BCL2 | 1.00 | 1.9 | 5.2 | AA291323 | ||
| Inhibitor of apoptosis 5 | 0.56 | 2.8 | 1.6 | 2.4 | 1.8 | AI972925 |
| caspase 6, apoptosis-related cysteine protease | 0.79 | 0.7 | 2.6 | 1.3 | 2.8 | W45688 |
| Apoptosis-related protein PNAS-1 | 0.46 | 2.2 | 1.4 | 2.3 | 2.9 | AA521316 |
| caspase 8, apoptosis-related cysteine protease | 0.95 | 2.2 | 1.0 | 0.6 | 2.0 | AA448468 |
[00107] Table 22: polynucleotide a downregulated by peptide treatment in a549 epithelial cells. Cationic peptides at a concentration of 50. mu.g/ml were found to reduce the expression of several polynucleotides. The peptides were incubated with human A549 epithelial cells for 4 hours, RNA was isolated, converted to labeled cDNA probes, and hybridized to human cDNA array ID # PRHU 03-S3. The intensity of the polynucleotide in the unstimulated cells is shown in the second column. "ratio peptide: by unstimulated "column is meant the intensity of polynucleotide expression in cells stimulated with the peptide divided by the intensity of the unstimulated cells.
| Polynucleotides/proteins | Is not coveredIntensity of stimulus | Ratiometric peptide to no stimulation | Registration number | |||
| ID 3 | ID 3 | ID 19 | ID 1 | |||
| TLR 1 | 3.22 | 0.35 | 0.31 | 0.14 | 0.19 | AI339155 |
| TLR 2 | 2.09 | 0.52 | 0.31 | 0.48 | 0.24 | T57791 |
| TLR 5 | 8.01 | 0.12 | 0.39 | N41021 | ||
| TLR 7 | 5.03 | 0.13 | 0.11 | 0.20 | 0.40 | N30597 |
| TNF receptor associated factor 2 | 0.82 | 1.22 | 0.45 | 2.50 | 2.64 | T55353 |
| TNF receptor associated factor 3 | 3.15 | 0.15 | 0.72 | 0.32 | AA504259 | |
| TNF receptor superfamily, member 12 | 4.17 | 0.59 | 0.24 | 0.02 | W71984 | |
| TNF R superfamily, member 17 | 2.62 | 0.38 | 0.55 | 0.34 | AA987627 | |
| TRAF and TNF receptor associated proteins | 1.33 | 0.75 | 0.22 | 0.67 | 0.80 | AA488650 |
| IL-1 receptor, type I | 1.39 | 0.34 | 0.72 | 1.19 | 0.34 | AA464526 |
| IL-2 receptor, alpha | 2.46 | 0.41 | 0.33 | 0.58 | AA903183 | |
| IL-2 receptor, gamma (severe combined immunodeficiency) | 3.34 | 0.30 | 0.24 | 0.48 | N54821 | |
| IL-12 receptor, beta 2 | 4.58 | 0.67 | 0.22 | AA977194 | ||
| IL-18 receptor 1 | 1.78 | 0.50 | 0.42 | 0.92 | 0.56 | AA482489 |
| TGF-beta receptor III | 2.42 | 0.91 | 0.24 | 0.41 | 0.41 | H62473 |
| Leukotriene b4 receptor (chemokine receptor-like-1) | 1.00 | 1.38 | 4.13 | 0.88 | AI982606 | |
| Small molecule inducible cytokine subfamily A (Cys-Cys), member 18 | 2.26 | 0.32 | 0.44 | 1.26 | AA495985 | |
| Small molecule inducible cytokine subfamily A (Cys-Cys), member 20 | 2.22 | 0.19 | 0.38 | 0.45 | 0.90 | AI285199 |
| Small molecule inducible cytokine subfamily A | 2.64 | 0.38 | 0.31 | 1.53 | AA916836 | |
| (Cys-Cys), member 23 | ||||||
| Small molecule inducible cytokine subfamily B (Cys-X-Cys), member 6 (granulocyte chemotactic protein 2) | 3.57 | 0.11 | 0.06 | 0.28 | 0.38 | AI889554 |
| Small molecule inducible cytokine subfamily B (Cys-X-Cys), member 10 | 2.02 | 0.50 | 1.07 | 0.29 | 0.40 | AA878880 |
| Small molecule inducible cytokine A3 (homologous to murine Mip-1 a) | 2.84 | 1.79 | 0.32 | 0.35 | AA677522 | |
| Cytokine-induced kinase | 2.70 | 0.41 | 0.37 | 0.37 | 0.34 | AA489234 |
| Complement component Clq receptors | 1.94 | 0.46 | 0.58 | 0.51 | 0.13 | AI761788 |
| Calnexin type 11, 2, OB-calnexin (osteoblasts) | 2.00 | 0.23 | 0.57 | 0.30 | 0.50 | AA136983 |
| Cadherin type 3, 1, P-cadherin (placenta) | 2.11 | 0.43 | 0.53 | 0.10 | 0.47 | AA425217 |
| Cadherin, EGF LAGseven-pass G receptor 2, flumingo homolog | 1.67 | 0.42 | 0.41 | 1.21 | 0.60 | H39187 |
| Calyosin 13, H-calyosin (heart) | 1.78 | 0.37 | 0.40 | 0.56 | 0.68 | R41787 |
| Lectin L (lymphocyte adhesion molecule 1) | 4.43 | 0.03 | 0.23 | 0.61 | H00662 | |
| Vascular cell adhesion molecule 1 | 1.40 | 0.20 | 0.72 | 0.77 | 0.40 | H16591 |
| Intermolecular adhesion molecule | 1.00 | 0.12 | 0.31 | 2.04 | 1.57 | AA479188 |
| 3 | ||||||
| Integrin,. alpha.1 | 2.42 | 0.41 | 0.26 | 0.56 | AA450324 | |
| Integrin,. alpha.7 | 2.53 | 0.57 | 0.39 | 0.22 | 0.31 | AA055979 |
| Integrin,. alpha.9 | 1.16 | 0.86 | 0.05 | 0.01 | 2.55 | AA865557 |
| Integrin,. alpha.10 | 1.00 | 0.33 | 0.18 | 1.33 | 2.55 | AA460959 |
| Integrin,. beta.5 | 1.00 | 0.32 | 1.52 | 1.90 | 0.06 | AA434397 |
| Integrin,. beta.8 | 3.27 | 0.10 | 1.14 | 0.31 | 0.24 | W56754 |
| Disintegrin and metalloprotease domain 18 | 2.50 | 0.40 | 0.29 | 0.57 | 0.17 | AI205675 |
| Disintegrin-like and metalloproteases possess the platelet binding protein 1 motif, 3 | 2.11 | 0.32 | 0.63 | 0.47 | 0.35 | AA398492 |
| Disintegrin-like and metalloproteases possess the platelet binding protein 1 motif, 5 | 1.62 | 0.39 | 0.42 | 1.02 | 0.62 | AI375048 |
| T cell receptor effector molecules | 1.00 | 0.41 | 1.24 | 1.41 | 0.45 | AI453185 |
| Diphtheria toxin receptor (heparin binding epidermal growth factor-like growth factor) | 1.62 | 0.49 | 0.85 | 0.62 | 0.15 | R45640 |
| Vasoactive intestinal peptide receptor 1 | 2.31 | 0.43 | 0.31 | 0.23 | 0.54 | H73241 |
| Fc fragment of IgG, low affinity IIIb, receptor (CD16) | 3.85 | -0.20 | 0.26 | 0.76 | 0.02 | H20822 |
| Fc fragment of IgG, low affinity IIb, receptor (CD32) | 1.63 | 0.27 | 0.06 | 1.21 | 0.62 | R68106 |
| The Fc fragment of IgE, high affinity I, receptor is; alpha polypeptides | 1.78 | 0.43 | 0.00 | 0.56 | 0.84 | AI676097 |
| Leukocyte immunoglobulin-like receptor, subfamily A | 2.25 | 0.44 | 0.05 | 0.38 | 0.99 | N63398 |
| Leukocyte immunoglobulin-like receptor, subfamily B (with TM and ITIM domains), member 3 | 14.21 | 1.10 | 0.07 | AI815229 | ||
| Leukocyte immunoglobulin-like receptor, subfamily B (with TM and ITIM domains), member 4 | 2.31 | 0.75 | 0.43 | 0.19 | 0.40 | AA076350 |
| Leukocyte immunoglobulin-like receptor, subfamily B | 1.67 | 0.35 | 0.60 | 0.18 | 0.90 | H54023 |
| Peroxisome proliferator activated receptor, alpha | 1.18 | 0.38 | 0.85 | 0.87 | 0.26 | AI739498 |
| Protein tyrosine phosphatase, receptor type, f polypeptide (PTPRF), interacting protein (liprin), alpha 1 | 2.19 | 0.43 | 1.06 | 0.46 | N49751 | |
| Protein tyrosine phosphatases, receptor types, C | 1.55 | 0.44 | 0.64 | 0.30 | 0.81 | H74265 |
| Protein tyrosine phosphatases, receptor types, E | 2.08 | 0.23 | 0.37 | 0.56 | 0.48 | AA464542 |
| Protein tyrosine phosphatases, receptor types, N polypeptide 2 | 2.27 | 0.02 | 0.44 | 0.64 | AA464590 | |
| Protein tyrosine phosphatase, receptor type, H | 2.34 | 0.11 | 0.43 | 0.24 | 0.89 | AI924306 |
| Protein tyrosine phosphatase, receptor type, Z polypeptide 1 | 1.59 | 0.63 | 0.34 | 0.72 | 0.35 | AA476461 |
| Protein tyrosine phosphatase, non-receptor type 21 | 1.07 | 0.94 | 0.43 | 0.25 | 1.13 | H03504 |
| MAP kinase 8 interacting protein 2 | 1.70 | 0.07 | 0.85 | 0.47 | 0.59 | AA418293 |
| MAP kinase | 1.27 | 0.37 | 0.79 | 1.59 | -5.28 | AA402447 |
| Enzyme 4 | ||||||
| MAP kinase 14 | 1.00 | 0.34 | 0.66 | 2.10 | 1.49 | W61116 |
| MAP kinase 8 interacting protein 2 | 2.90 | 0.16 | 0.35 | 0.24 | 0.55 | AI202738 |
| MAP kinase 12 | 1.48 | 0.20 | 0.91 | 0.58 | 0.68 | AA053674 |
| MAP kinase 3 | 2.21 | 0.45 | 0.20 | 1.03 | 0.41 | AA043537 |
| MAP kinase 6 | 2.62 | 0.37 | 0.38 | 0.70 | AW084649 | |
| MAP kinase 4 | 1.04 | 0.96 | 0.09 | 0.29 | 2.79 | AA417711 |
| MAP kinase 11 | 1.53 | 0.65 | 0.41 | 0.99 | 0.44 | R80779 |
| MAP kinase 10 | 1.32 | 1.23 | 0.27 | 0.50 | 0.76 | H01340 |
| MAP kinase 9 | 2.54 | 0.57 | 0.39 | 0.16 | 0.38 | AA157286 |
| MAP kinase 1 | 1.23 | 0.61 | 0.42 | 0.81 | 1.07 | AI538525 |
| MAP kinase 8 | 0.66 | 1.52 | 1.82 | 9.50 | 0.59 | W56266 |
| MAP kinase activated protein kinase 3 | 0.52 | 2.13 | 2.68 | 1.13 | 1.93 | W68281 |
| MAP kinase 2 | 0.84 | 1.20 | 3.35 | 0.02 | 1.31 | AA425826 |
| MAP kinase 7 | 1.00 | 0.97 | 1.62 | 7.46 | AA460969 | |
| MAP kinase 7 | 0.09 | 11.45 | 11.80 | 33.43 | H39192 | |
| MAP kinase 6 | 0.10 | 17.83 | 9.61 | 32.30 | H07920 | |
| Regulatory protein 5 for G protein signaling | 3.7397 | 0.27 | 0.06 | 0.68 | 0.18 | AA668470 |
| Regulatory protein 13 of G protein signaling | 1.8564 | 0.54 | 0.45 | 0.07 | 1.09 | H70047 |
| G protein-coupled receptors | 1.04 | 1.84 | 0.16 | 0.09 | 0.96 | R91916 |
| G protein-coupled receptor | 1.78 | 0.32 | 0.56 | 0.39 | 0.77 | AI953187 |
| Body 17 | ||||||
| G proteinCoupled receptor kinase 7 | 2.62 | 0.34 | 0.91 | 0.38 | AA488413 | |
| Seven transmembrane receptors in solitary son, associated with chemokines | 7.16 | 1.06 | 0.10 | 0.11 | 0.14 | AI131555 |
| Apoptosis-antagonistic transcription factors | 1.00 | 0.28 | 2.50 | 1.28 | 0.19 | AI439571 |
| caspase 1, apoptosis-related cysteine protease (interleukin 1, beta, convertase) | 2.83 | 0.44 | 0.33 | 0.35 | T95052 | |
| Programmed cell death 8 (factor inducing apoptosis) | 1.00 | 1.07 | 0.35 | 1.94 | 0.08 | AA496348 |
[00108] Table 23: a549 cells are a pro-inflammatory polynucleotide that is upregulated by peptide treatment. Cationic peptide concentrations of 50. mu.g/ml were found to increase the expression of certain pro-inflammatory polynucleotides (data are part of Table 21). The peptides were incubated with human A549 epithelial cells for 4 hours, RNA was isolated, converted to labeled cDNA probes, and hybridized to human cDNA array ID # PRHU 03-S3. The intensity of the polynucleotide in the unstimulated cells is shown in the second column. "ratio peptide: by unstimulated "column is meant the intensity of polynucleotide expression in cells stimulated with the peptide divided by the intensity of the unstimulated cells.
| Polynucleotides/proteins and functions | Strength of not being stimulated | Ratiometric peptide to no stimulation | Registration number | |||
| ID 2 | ID 3 | ID 19 | ID 1 | |||
| IL-11R α; receptor of proinflammatory cytokines, inflammation | 0.55 | 2.39 | 0.98 | 4.85 | 1.82 | AA454657 |
| IL-17R; IL-17 receptor, inducer of cytokine production in epithelial cells | 0.54 | 2.05 | 1.97 | 1.52 | 1.86 | AW029299 |
| Small molecule inducible cytokine subfamily a, member 21; chemotactic factor | 1.00 | 3.88 | 2.41 | AI922341 | ||
| CD 31; leukocyte and cell-to-cell adhesion (PECAM) | 0.59 | 2.71 | 3.13 | 1.01 | 1.68 | R22412 |
| CCR 6; receptors for the cytokine MIP-3 alpha | 0.14 | 4.51 | 7.75 | 6.92 | 7.79 | N57964 |
| Integrin, α 2(CD49B, the α 2 subunit of the VLA-2 receptor); adhesion to leukocytes | 1.00 | 0.89 | 2.44 | 3.62 | 0.88 | AA463257 |
| Integrin, α 3(CD49C, the α 3 subunit of the VLA-3 receptor); adhesion to leukocytes | 0.94 | 0.79 | 2.51 | 1.88 | 1.07 | AA424695 |
| Integrin, α E; adhesion | 0.01 | 179.33 | 120.12 | 28.48 | 81.37 | AA425451 |
| Integrin, β 4; leukocyte adhesion | 0.65 | 0.79 | 2.17 | 4.94 | 1.55 | AA485668 |
| C-type lectin-like receptor 2; leukocyte adhesion | 0.45 | 2.09 | 7.92 | 2.24 | 5.29 | H70491 |
[00109] Table 24: a pro-inflammatory polynucleotide that is down-regulated by peptide treatment in a549 cells. Cationic peptide concentrations of 50. mu.g/ml were found to reduce the expression of certain pro-inflammatory polynucleotides (data is part of Table 22). The peptides were incubated with human A549 epithelial cells for 4 hours, RNA was isolated, converted to labeled cDNA probes, and hybridized to human cDNA array ID # PRHU 03-S3. The intensity of the polynucleotide in the unstimulated cells is shown in the second column. The column "ratio peptide: unstimulated" refers to the intensity of polynucleotide expression in cells stimulated with the peptide divided by the intensity of unstimulated cells.
| A polynucleotide/protein; function(s) | Strength of not being stimulated | Ratiometric peptide to no stimulation | Registration number | |||
| ID 2 | ID 3 | ID 19 | ID 1 | |||
| Toll-like receptor (TLR) 1; leather pair | 3.22 | 0.35 | 0.31 | 0.14 | 0.19 | AI339155 |
| Response by Lane-positive bacteria | ||||||
| TLR 2; response to gram-Positive bacteria and Yeast | 2.09 | 0.52 | 0.31 | 0.48 | 0.24 | T57791 |
| TLR 5; can enhance the responsiveness of other TLRs to flagellin | 8.01 | 0.12 | 0.39 | N41021 | ||
| TLR 7; derived host defense mechanisms | 5.03 | 0.13 | 0.11 | 0.20 | 0.40 | N30597 |
| TNF receptor-related factor 2; inflammation(s) | 0.82 | 1.22 | 0.45 | 2.50 | 2.64 | T55353 |
| TNF receptor-related factor 3; inflammation(s) | 3.15 | 0.15 | 0.72 | 0.32 | AA504259 | |
| TNF receptor superfamily, member 12; inflammation(s) | 4.17 | 0.59 | 0.24 | 0.02 | W71984 | |
| TNF receptor superfamily, member 17; inflammation(s) | 2.62 | 0.38 | 0.55 | 0.34 | AA987627 | |
| TRAF and TNF receptor associated proteins; TNF signaling | 1.33 | 0.75 | 0.22 | 0.67 | 0.80 | AA488650 |
| Small molecule inducible cytokine subfamily a, member 18; chemotactic factor | 2.26 | 0.32 | 0.44 | 1.26 | AA495985 | |
| Small molecule inducible cytokine subfamily a, member 20; chemotactic factor | 2.22 | 0.19 | 0.38 | 0.45 | 0.90 | AI285199 |
| Small molecule inducible cytokine subfamily a, member 23; chemotactic factor | 2.64 | 0.38 | 0.31 | 1.53 | AA916836 | |
| Small molecule induced cytokine subfamily | 3.57 | 0.11 | 0.06 | 0.28 | 0.38 | AI889554 |
| Family B, member 6; (granulocyte chemotactic protein); chemotactic factor | ||||||
| Small molecule inducible cytokine subfamily B, member 10; chemotactic factor | 2.02 | 0.50 | 1.07 | 0.29 | 0.40 | AA878880 |
| Small molecule inducible cytokine a3 (homologous to murine Mip-1 a); chemotactic factor | 2.84 | 1.79 | 0.32 | 0.35 | AA677522 | |
| IL-12 receptor, β 2; interleukin and interferon receptors | 4.58 | 0.67 | 0.22 | AA977194 | ||
| IL-18 receptor 1; induction of INF-gamma | 1.78 | 0.50 | 0.42 | 0.92 | 0.56 | AA482489 |
| Lectin L (leukocyte adhesion molecule 1); leukocyte adhesion | 4.43 | 0.03 | 0.23 | 0.61 | H00662 | |
| Vascular cell adhesion molecule 1; leukocyte adhesion | 1.40 | 0.20 | 0.72 | 0.77 | 0.40 | H16591 |
| Intercellular adhesion molecule 3; leukocyte adhesion | 1.00 | 0.12 | 0.31 | 2.04 | 1.57 | AA479188 |
| Integrin, α 1; leukocyte adhesion | 2.42 | 0.41 | 0.26 | 0.56 | AA450324 |
[00110] Table 25: an anti-inflammatory polynucleotide that is upregulated by peptide treatment in a549 cells. Cationic peptide concentrations of 50. mu.g/ml were found to increase the expression of certain anti-inflammatory polynucleotides (data are part of Table 21). The peptides were incubated with human A549 epithelial cells for 4 hours, RNA was isolated, converted to labeled cDNA probes, and hybridized to human cDNA array ID # PRHU 03-S3. The intensity of the polynucleotide in the unstimulated cells is shown in the second column. The column "ratio peptide: unstimulated" refers to the intensity of polynucleotide expression in cells stimulated with the peptide divided by the intensity of unstimulated cells.
| A polynucleotide/protein; function(s) | Strength of not being stimulated | Ratio peptide: is not stimulated | Registration number | |||
| ID 2 | ID 3 | ID 19 | ID 1 | |||
| IL-1R antagonist homolog 1; inhibitors of septic shock | 0.00 | 3085.96 | 1855.90 | 869.57 | AI167887 | |
| IL-10R β; receptors for inhibitors of cytokine synthesis | 0.53 | 2.51 | 1.56 | 1.88 | 3.10 | AA486393 |
| TNF R, member 1B; apoptosis of cells | 0.28 | 17.09 | 3.01 | 14.93 | 3.60 | AA150416 |
| TNF R, member 5; apoptosis (CD40L) | 33.71 | 2.98 | 0.02 | H98636 | ||
| TNF R, member 11 b; apoptosis of cells | 1.00 | 5.29 | 4.50 | 0.78 | AA194983 | |
| IK cytokines, down regulators of HLA II; inhibition of antigen presentation | 0.50 | 3.11 | 2.01 | 1.74 | 3.29 | R39227 |
| TGFB inducible early growth response protein 2; anti-inflammatory cytokines | 0.90 | 2.38 | 2.08 | 0.87 | 1.11 | AI473938 |
| CD 2; adhesion molecules, binding to LFAp3 | 1.00 | 2.62 | 0.87 | 1.15 | 0.88 | AA927710 |
[00111] Table 26: an anti-inflammatory polynucleotide that is down-regulated by peptide treatment in a549 cells. Cationic peptide concentrations of 50. mu.g/ml were found to reduce the expression of certain anti-inflammatory polynucleotides (data are part of Table 21). The peptides were incubated with human A549 epithelial cells for 4 hours, RNA was isolated, converted to labeled cDNA probes, and hybridized to human cDNA array ID # PRHU 03-S3. The intensity of the polynucleotide in the unstimulated cells is shown in the second column. The column "ratio peptide: unstimulated" refers to the intensity of polynucleotide expression in cells stimulated with the peptide divided by the intensity of unstimulated cells.
| A polynucleotide/protein; function(s) | Strength of not being stimulated | Ratiometric peptide to no stimulation | Registration number | |||
| ID 2 | ID 3 | ID 19 | ID 1 | |||
| MAP kinase 9 | 2.54 | 0.57 | 0.39 | 0.16 | 0.38 | AA157286 |
[00112] Table 27: primary human macrophages are encoded by SEQ ID NO: 6 an up-regulated polynucleotide. The peptide SEQ ID NO: 6 increases the expression of a plurality of polynucleotides. Peptides were incubated with Human macrophages for 4 hours, RNA was isolated, converted to labeled cDNA probes, and hybridized to a Human Operon array (PRHU 04). The intensity of the polynucleotide in the unstimulated cells is shown in the second column. "ratio peptide treatment-control" column refers to the intensity of polynucleotide expression in cells stimulated with peptide divided by the intensity of non-stimulated cells.
| Gene (registration number) | Comparison: unstimulated cells | Ratio peptide treatment: control |
| Proteoglycan 2(Z26248) | 0.69 | 9.3 |
| Unknown (AK001843) | 26.3 | 8.2 |
| Phosphorylase kinase alpha 1(X73874) | 0.65 | 7.1 |
| Actin, α 3(M86407) | 0.93 | 6.9 |
| DKFZP586B2420 protein (AL050143) | 0.84 | 5.9 |
| Unknown (AL109678) | 0.55 | 5.6 |
| Transcription factor 21(AF047419) | 0.55 | 5.4 |
| Unknown (A433612) | 0.62 | 5.0 |
| Chromosome condensation 1-sample (AF060219) | 0.69 | 4.8 |
| Unknown (AL137715) | 0.66 | 4.4 |
| Inhibitor of apoptosis 4(U75285) | 0.55 | 4.2 |
| Nuclear factor 2 (NM-012461) acting with TERF1(TRF1) | 0.73 | 4.2 |
| LINE reverse transcription rotary seat element 1(M22333) | 6.21 | 4.0 |
| 1-acylglycerol-3-phosphate O-acyltransferase 1(U56417) | 0.89 | 4.0 |
| Vacuolar protic atpase, subunit D; V-ATPase, subunit D (X71490) | 1.74 | 4.0 |
| KIAA0592 protein (AB011164) | 0.70 | 4.0 |
| KQT-like subfamily member 4 of voltage-gated potassium channel (AF105202) | 0.59 | 3.9 |
| CDC14 homolog A (AF000367) | 0.87 | 3.8 |
| Histone folding protein CHRAC17(AF070640) | 0.63 | 3.8 |
| Cryptogamic pigment 1(D83702) | 0.69 | 3.8 |
| Pancreatic zymogen granule membrane associated protein (AB035541) | 0.71 | 3.7 |
| Sp3 transcription factor (X68560) | 0.67 | 3.6 |
| Hypothetical protein FLJ20495(AK000502) | 0.67 | 3.5 |
| E2F transcription factor 5, p130 binding (U31556) | 0.56 | 3.5 |
| Hypothetical protein FLJ20070(AK000077) | 1.35 | 3.4 |
| Glycoprotein IX (X52997) | 0.68 | 3.4 |
| KIAA1013 protein (AB023230) | 0.80 | 3.4 |
| Eukaryotic translation initiation factor 4A, isoform 2(AL137681) | 2.02 | 3.4 |
| FYN binding protein (AF198052) | 1.04 | 3.3 |
| Guanylate binding protein, gamma-transducing active polypeptide 1(U41492) | 0.80 | 3.3 |
| Glypican 1(X54232) | 0.74 | 3.2 |
| Mucosal vascular addressen cell adhesion molecule 1(U43628) | 0.65 | 3.2 |
| Lymphocyte antigen (M38056) | 0.70 | 3.2 |
| H1 Histone family, member 4(M60748) | 0.81 | 3.0 |
| Translation inhibiting protein p14.5(X95384) | 0.78 | 3.0 |
| Hypothetical protein FLJ20689(AB032978) | 1.03 | 2.9 |
| KIAA1278 protein (AB03104) | 0.80 | 2.9 |
| Unknown (AL031864) | 0.95 | 2.9 |
| Chymotrypsin-like protease (X71877) | 3.39 | 2.9 |
| Network cavity calcium binding protein (NM _001219) | 2.08 | 2.9 |
| Protein kinase, cAMP dependent, regulatory, type I, beta (M65066) | 7.16 | 2.9 |
| POU Domain, class 4, transcription factor 2(U06233) | 0.79 | 2.8 |
| POU Domain, class 2, correlation factor 1(Z49194) | 1.09 | 2.8 |
| KIAA0532 protein (AB011104) | 0.84 | 2.8 |
| Not known (AF068289) | 1.01 | 2.8 |
| Unknown (AL117643) | 0.86 | 2.7 |
| Cathepsin E (M84424) | 15.33 | 2.7 |
| Interstitial metalloprotease 23A (AF056200) | 0.73 | 2.7 |
| Interferon receptor 2(L42243) | 0.70 | 2.5 |
| MAP kinase 1(L11284) | 0.61 | 2.4 |
| Protein kinase C, alpha (X52479) | 0.76 | 2.4 |
| c-Cb1 action protein (AF230904) | 0.95 | 2.4 |
| c-fos inducible growth factor (Y12864) | 0.67 | 2.3 |
| Cyclin-dependent kinase inhibitor 1B (S76988) | 0.89 | 2.2 |
| Zinc finger protein 266(X78924) | 1.67 | 2.2 |
| MAP kinase 14(L35263) | 1.21 | 2.2 |
| KIAA0922 protein (AB023139) | 0.96 | 2.1 |
| Bone morphogenetic protein 1(NM _006129) | 1.10 | 2.1 |
| NADH dehydrogenase 1. alpha. sub-complex, 10(AF087661) | 1.47 | 2.1 |
| Bone morphogenetic protein receptor, type 1B (U89326) | 0.50 | 2.1 |
| Interferon regulatory factor 2(NM002199) | 1.46 | 2.0 |
| Protease, serine, 21(AB031331) | 0.89 | 2.0 |
[00113] Table 28: primary human macrophages are encoded by SEQ ID NO: 6 down-regulated polynucleotide. The peptide SEQ ID NO: 6 reduces the expression of a plurality of polynucleotides. Peptides were incubated with Human macrophages for 4 hours, RNA was isolated, converted to labeled cDNA probes, and hybridized to a Human Operon array (PRHU 04). The intensity of the polynucleotide in the unstimulated cells is shown in the second column. "ratio peptide treatment: control "column" refers to the intensity of polynucleotide expression in cells stimulated with peptide divided by the intensity of non-stimulated cells.
| Gene (registration number) | Comparison: unstimulated cells | Ratio peptide treatment: control |
| Unknown (AL049263) | 17 | 0.06 |
| Integrin associated kinase (U40282) | 2.0 | 0.13 |
| KIAA0842 protein (AB020649) | 1.1 | 0.13 |
| Unknown (AB037838) | 13 | 0.14 |
| Granulin (AF055008) | 8.6 | 0.14 |
| Glutathione peroxidase 3(NM _002084) | 1.2 | 0.15 |
| KIAA0152 Gene product (D63486) | 0.9 | 0.17 |
| TGFB 1-inducible anti-apoptotic factor 1(D86790) | 0.9 | 0.19 |
| Disintegrin protease (Y13323) | 1.5 | 0.21 |
| Proteasome subunit beta type 7(D38048) | 0.7 | 0.22 |
| Sp1 accessory factor required for transcriptional activation of subunit 3(AB 033042) | 0.9 | 0.23 |
| TNF receptor superfamily, member 14(U81232) | 0.8 | 0.26 |
| Proteasome 26S subunit non-atpase 8 (D38047)) | 1.1 | 0.28 |
| Proteasome subunit beta type, 4(D26600) | 0.7 | 0.29 |
| TNF receptor superfamily member 1B (M32315) | 1.7 | 0.29 |
| Cytochrome c oxidase subunit Vic (X13238) | 3.3 | 0.30 |
| S100 calcium binding protein A4(M80563) | 3.8 | 0.31 |
| Proteasome subunit alpha type, 6(X59417) | 2.9 | 0.31 |
| Proteasome 26S subunit non-ATPase, 10(AL031177) | 1.0 | 0.32 |
| MAP kinase 2(NM _006609) | 0.8 | 0.32 |
| Ribosomal protein L11(X79234) | 5.5 | 0.32 |
| Interstitial metalloprotease 14(Z48481) | 1.0 | 0.32 |
| Proteasome subunit beta type, 5(D29011) | 1.5 | 0.33 |
| MAP kinase activated protein kinase 2: (U12779) | 1.5 | 0.34 |
| caspase 3(U13737) | 0.5 | 0.35 |
| jun D protooncogene (X56681) | 3.0 | 0.35 |
| Proteasome 26S subunit, ATPase, 3(M34079) | 1.3 | 0.35 |
| IL-1 receptor like 1(AB012701) | 0.7 | 0.35 |
| Interferon alpha inducibility protein (AB019565) | 13 | 0.35 |
| SDF receptor 1 (NM-012428) | 1.6 | 0.35 |
| Cathepsin D (M63138) | 46 | 0.36 |
| MAP kinase 3(D87116) | 7.4 | 0.37 |
| TGF, beta inducibility (M77349) | 1.8 | 0.37 |
| TNF receptor superfamily, member 10b (AF016266) | 1.1 | 0.37 |
| Proteasome subunit beta type, 6(M34079) | 1.3 | 0.38 |
| Nuclear receptor binding protein (NM _013392) | 5.2 | 0.38 |
| Unknown (AL050370) | 1.3 | 0.38 |
| Protease inhibitor 1 alpha-1-antitrypsin (X01683) | 0.7 | 0.40 |
| Proteasome subunit alpha type, 7(AF054185) | 5.6 | 0.40 |
| TNF-alpha factor induced by LPS (NM-004862) | 5.3 | 0.41 |
| Transferrin receptor (X01060) | 14 | 0.42 |
| Proteasome 26S subunit non-ATP enzyme 13(AB009398) | 1.8 | 0.44 |
| MAP kinase 5(U25265) | 1.3 | 0.44 |
| Tissue protein L (X12451) | 15 | 0.44 |
| IL-1 receptor associated kinase 1(L76191) | 1.7 | 0.45 |
| MAP kinase 2(U07349) | 1.1 | 0.46 |
| Peroxisome proliferator activated receptor delta (AL022721) | 2.2 | 0.46 |
| TNF superfamily, member 15(AF039390) | 16 | 0.46 |
| Cell death defense 1(D15057) | 3.9 | 0.46 |
| TNF superfamily member 10(U37518) | 287 | 0.46 |
| Histon H (X16832) | 14 | 0.47 |
| Protease inhibitor 12(Z81326) | 0.6 | 0.48 |
| Proteasome subunit alpha type, 4(D00763) | 2.6 | 0.49 |
| Proteasome 26S subunit ATPase, 1(L02426) | 1.8 | 0.49 |
| Proteasome 26S subunit ATPase, 2(D11094) | 2.1 | 0.49 |
| caspase 7(U67319) | 2.4 | 0.49 |
| Interstitial metalloprotease 7(Z11887) | 2.5 | 0.49 |
[00114] Table 29: HBE cells are encoded by SEQ ID NO: 1 an up-regulated polynucleotide. The peptide SEQ ID NO: 1 increases the expression of a plurality of polynucleotides. Peptides were incubated with Human HBE epithelial cells for 4 hours, RNA was isolated, converted to labeled cDNA probes, and hybridized to a Human Operon array (PRHU 04). The intensity of the polynucleotide in the unstimulated cells is shown in the third column. "ratio peptide treatment: control "column" refers to the intensity of polynucleotide expression in cells stimulated with peptide divided by the intensity of non-stimulated cells.
| Registration number | Gene | Comparison: unstimulated cells | Ratio peptide treatment: control |
| AL110161 | Is unknown | 0.22 | 5218.3 |
| AF131842 | Is unknown | 0.01 | 573.1 |
| AJ000730 | Family of solute carriers | 0.01 | 282.0 |
| Z25884 | Chloride ion channel 1 | 0.01 | 256.2 |
| M93426 | Protein tyrosine phosphatase receptor-type, zeta | 0.01 | 248.7 |
| X65857 | Olfactory receptor, family 1, subfamily D, member 2 | 0.01 | 228.7 |
| M55654 | TATA sequence box binding proteins | 0.21 | 81.9 |
| AK001411 | Hypothetical proteins | 0.19 | 56.1 |
| D29643 | Polyperpene diphospho oligosaccharide-proteoglycan base | 1.56 | 55.4 |
| Transferases | |||
| AF006822 | Myelin transcription factor 2 | 0.07 | 55.3 |
| AL117601 | Is unknown | 0.05 | 53.8 |
| AL117629 | DKFZP434C245 protein | 0.38 | 45.8 |
| M59465 | Tumor necrosis factor, alpha-inducing protein 3 | 0.50 | 45.1 |
| AB013456 | Aquaporin 8 | 0.06 | 41.3 |
| AJ131244 | SEC 24-related Gene family, Member A | 0.56 | 25.1 |
| AL110179 | Is unknown | 0.87 | 24.8 |
| AB037844 | Is unknown | 1.47 | 20.6 |
| Z47727 | Polymerase II polypeptide K | 0.11 | 20.5 |
| AL035694 | Is unknown | 0.81 | 20.4 |
| X68994 | Human CREB gene | 0.13 | 19.3 |
| AJ238379 | Hypothetical proteins | 1.39 | 18.5 |
| NM_003519 | H2B histone family member | 0.13 | 18.3 |
| U16126 | Glutamate receptor, ionotropic kainate2 | 0.13 | 17.9 |
| U29926 | Adenosine monophosphate deaminase | 0.16 | 16.3 |
| AK001160 | Hypothetical proteins | 0.39 | 14.4 |
| U18018 | ets variant gene 4 | 0.21 | 12.9 |
| D80006 | KIAA0184 protein | 0.21 | 12.6 |
| AK000768 | Hypothetical proteins | 0.30 | 12.3 |
| X99894 | Insulin promoter factor 1 | 0.26 | 12.0 |
| AL031177 | Is unknown | 1.09 | 11.2 |
| AF052091 | Is unknown | 0.28 | 10.9 |
| L38928 | 5, 10-methylene tetrahydrofolate synthetase | 0.22 | 10.6 |
| AL117421 | Is unknown | 0.89 | 10.1 |
| AL133606 | Hypothetical proteins | 0.89 | 9.8 |
| NM_016227 | Membrane protein CH1 | 0.28 | 9.6 |
| NM_006594 | Connexin-related protein complex 4 | 0.39 | 9.3 |
| U54996 | ZW10 homologue, protein | 0.59 | 9.3 |
| AJ007557 | Potassium channels | 0.28 | 9.0 |
| AF043938 | Muscle RAS oncogene | 1.24 | 8.8 |
| AK001607 | Is unknown | 2.74 | 8.7 |
| AL031320 | Peroxisome biogenesis factor | 0.31 | 8.4 |
| Son 30 | |||
| D38024 | Is unknown | 0.31 | 8.3 |
| AF059575 | LIM homeobox TF | 2.08 | 8.2 |
| AF043724 | Hepatitis a virus cell receptor 1 | 0.39 | 8.1 |
| AK002062 | Hypothetical proteins | 2.03 | 8.0 |
| L13436 | Natriuretic polypeptide receptors | 0.53 | 7.8 |
| U33749 | Thyroid transcription factor 1 | 0.36 | 7.6 |
| AF011792 | Cell cycle process 2 protein | 0.31 | 7.6 |
| AK000193 | Hypothetical proteins | 1.18 | 6.8 |
| AF039022 | Exportin (tRNA) | 0.35 | 6.8 |
| M17017 | Interleukin 8 | 0.50 | 6.7 |
| AF044958 | NADH dehydrogenase | 0.97 | 6.5 |
| U35246 | Vesicle sorting proteins | 0.48 | 6.5 |
| AK001326 | Tetratransmembrane protein 3 | 1.59 | 6.5 |
| M55422 | Kruepe-related zinc finger proteins | 0.34 | 6.4 |
| U44772 | Palmitoyl-protein thioesterases | 1.17 | 6.3 |
| AL117485 | Hypothetical proteins | 0.67 | 5.9 |
| AB037776 | Is unknown | 0.75 | 5.7 |
| AF131827 | Is unknown | 0.69 | 5.6 |
| AL137560 | Is unknown | 0.48 | 5.2 |
| X05908 | Annexin A1 | 0.81 | 5.1 |
| X68264 | Melanoma adhesion molecules | 0.64 | 5.0 |
| AL161995 | neurturin | 0.86 | 4.9 |
| AF037372 | Cytochrome c oxidase | 0.48 | 4.8 |
| NM_016187 | Bridged integrin 2 | 0.65 | 4.8 |
| AL137758 | Is unknown | 0.57 | 4.8 |
| U59863 | TRAF family member-associated NFKB activators | 0.46 | 4.7 |
| Z30643 | Chloride ion channel Ka | 0.70 | 4.7 |
| D16294 | acetyl-CoA acyltransferase 2 | 1.07 | 4.6 |
| AJ132592 | Zinc finger protein 281 | 0.55 | 4.6 |
| X82324 | POU Domain TF | 1.73 | 4.5 |
| NM_016047 | CGI-110 protein | 1.95 | 4.5 |
| AK001371 | Hypothetical proteins | 0.49 | 4.5 |
| M60746 | H3 Histone family member D | 3.05 | 4.5 |
| AB033071 | Hypothetical proteins | 4.47 | 4.4 |
| AB002305 | KIAA0307 gene product | 1.37 | 4.4 |
| X92689 | UDP-N-acetyl-alpha-D-galacton | 0.99 | 4.4 |
| Sugar amine: polypeptide N-acetylgalactosamine aminotransferase | |||
| AL049543 | Glutathione peroxidase 5 | 1.62 | 4.3 |
| U43148 | PTCH(patched homolog) | 0.96 | 4.3 |
| M67439 | Dopamine receptor D5 | 2.61 | 4.2 |
| U09850 | Zinc finger protein 143 | 0.56 | 4.2 |
| L20316 | Glucagon receptor | 0.75 | 4.2 |
| AB037767 | Disintegrin-like and metalloproteases | 0.69 | 4.2 |
| NM_017433 | Myosin IIIA | 99.20 | 4.2 |
| D26579 | Disintegrin and metalloprotease domain 8 | 0.59 | 4.1 |
| L10333 | reticulon 1 | 1.81 | 4.1 |
| AK000761 | Is unknown | 1.87 | 4.1 |
| U91540 | NK homeobox family 3, A | 0.80 | 4.1 |
| Z17227 | Interleukin 10 receptor, beta | 0.75 | 4.0 |
[00115] Table 30: HBE cells were encoded by the peptide SEQ ID NO: 1 (50. mu.g/ml) down-regulated polynucleotide. The peptide SEQ ID NO: 1 reduces the expression of a plurality of polynucleotides. Peptides were incubated with Human HBE epithelial cells for 4 hours, RNA was isolated, converted to labeled cDNA probes, and hybridized to a Human Operon array (PRHU 04). The intensity of the polynucleotide in the unstimulated cells is shown in the third column. "ratio peptide treatment: control "column" refers to the intensity of polynucleotide expression in cells stimulated with peptide divided by the intensity of non-stimulated cells.
| Registration number | Gene | Comparison: unstimulated cells | Ratio peptide treatment: control |
| AC004908 | Is unknown | 32.4 | 0.09 |
| S70622 | G1 stage specific gene | 43.1 | 0.10 |
| Z97056 | DEAD/H box polypeptide | 12.8 | 0.11 |
| AK002056 | Hypothetical proteins | 11.4 | 0.12 |
| L33930 | CD24 antigen | 28.7 | 0.13 |
| X77584 | Thioredoxin | 11.7 | 0.13 |
| NM_014106 | PRO1914 protein | 25.0 | 0.14 |
| M37583 | H2A histone family member | 22.2 | 0.14 |
| U89387 | Polymerase (RNA) II polypeptide D | 10.2 | 0.14 |
| D25274 | ras-related botulinum toxin C3 | 10.3 | 0.15 |
| Substrate 1 | |||
| J04173 | Phosphoglyceride mutase 1 | 11.4 | 0.15 |
| U19765 | Zinc finger protein 9 | 8.9 | 0.16 |
| X67951 | Proliferation-related gene A | 14.1 | 0.16 |
| AL096719 | Actin inhibitory protein 2 | 20.0 | 0.16 |
| AF165217 | Promyoglobin 4 | 14.6 | 0.16 |
| NM_014341 | Mitochondrial Carrier protein homolog 1 | 11.1 | 0.16 |
| AL022068 | Is unknown | 73.6 | 0.17 |
| X69150 | Ribosomal protein S18 | 42.8 | 0.17 |
| AL031577 | Is unknown | 35.0 | 0.17 |
| AL031281 | Is unknown | 8.9 | 0.17 |
| AF090094 | Human ornithine decarboxylase-resistant mRNA | 10.3 | 0.17 |
| AL022723 | `HLA-GHistocompatibility antigens, class I, G | 20.6 | 0.18 |
| U09813 | ATP synthase, H + transporting mitochondrial F0 complex | 9.8 | 0.18 |
| AF000560 | Human TTF-1 interacting polypeptide 20 | 20.2 | 0.19 |
| NM_016094 | HSPC042 protein | 67.2 | 0.19 |
| AF047183 | NADH dehydrogenase | 7.5 | 0.19 |
| D14662 | Antioxidant protein 2 (non-selenium glutathione peroxidase, acid calcium dependent phospholipase) | 8.1 | 0.19 |
| X16662 | Annexin A8 | 8.5 | 0.19 |
| U14588 | Pilin | 11.3 | 0.19 |
| AL117654 | DKFZP586D0624 protein | 12.6 | 0.20 |
| AK001962 | Hypothetical proteins | 7.7 | 0.20 |
| L41559 | 6-pyruvoyl tetrahydropterin synthase/hepatocyte nuclear factor 1 alpha dimerizing cofactor | 9.1 | 0.20 |
| NM_016139 | 16.7Kd protein | 21.0 | 0.21 |
| NM_016080 | CGI-150 protein | 10.7 | 0.21 |
| U86782 | 26S proteasome-associated pad1 homolog | 6.7 | 0.21 |
| AJ400717 | Tumor protein, translation control protein 1 | 9.8 | 0.21 |
| X07495 | Homology box C4 | 31.0 | 0.21 |
| AL034410 | Is unknown | 7.3 | 0.22 |
| X14787 | Platelet binding protein 1 | 26.2 | 0.22 |
| AF081192 | Purine element-rich binding protein B | 6.8 | 0.22 |
| D49489 | Protein disulfide isomerase-related protein | 11.0 | 0.22 |
| NM_014051 | PTD011 protein | 9.3 | 0.22 |
| AK001536 | Is unknown | 98.0 | 0.22 |
| X62534 | 2 group of high-speed swimming proteins | 9.5 | 0.22 |
| AJ005259 | Endothelial differentiation related factor 1 | 6.7 | 0.22 |
| NM_000120 | Epoxide hydrolase 1, microsomal | 10.0 | 0.22 |
| M38591 | S100 calcium binding protein A10 | 23.9 | 0.23 |
| AF071596 | Immediate early corresponding protein 2 | 11.5 | 0.23 |
| X16396 | Methylenetetrahydrofolate dehydrogenase | 8.3 | 0.23 |
| AK000934 | ATP enzyme inhibitor precursors | 7.6 | 0.23 |
| AL117612 | Is unknown | 10.7 | 0.23 |
| AF119043 | Transcription intermediate factor 1 gamma | 7.3 | 0.23 |
| AF037066 | Solute carrier family 22 member 1-like antisense | 7.6 | 0.23 |
| AF134406 | Cytochrome c oxidase subunit | 13.3 | 0.23 |
| AE000661 | Is unknown | 9.2 | 0.24 |
| AL157424 | synaptojanin 2 | 7.2 | 0.24 |
| X56468 | Tyrosine 3-monooxygenase/tryptophan 5-monooxygenase activating protein | 7.2 | 0.24 |
| U39318 | Ubiquitin conjugating enzyme E2D3 | 10.7 | 0.24 |
| AL034348 | Is unknown | 24.4 | 0.24 |
| D26600 | Proteasome subunit beta type 4 | 11.4 | 0.24 |
| AB032987 | Is unknown | 16.7 | 0.24 |
| J04182 | Lysosomal associated membrane protein 1 | 7.4 | 0.24 |
| X78925 | Zinc finger protein 267 | 16.1 | 0.25 |
| NM_000805 | Gastrin | 38.1 | 0.25 |
| U29700 | Anti-muir hormone receptor, type II | 12.0 | 0.25 |
| Z98200 | Is unknown | 13.4 | 0.25 |
| U07857 | Signal recognition particles | 10.3 | 0.25 |
| L05096 | Human ribosomal protein L39 | 25.3 | 0.25 |
| AK001443 | Hypothetical proteins | 7.5 | 0.25 |
| K03515 | Phosphoglucose isomerase | 6.2 | 0.25 |
| X57352 | Interferon-inducible transmembrane protein 3 | 7.5 | 0.26 |
| J02883 | Pancreatic co-lipase | 5.7 | 0.26 |
| M24069 | Cold shock domain proteins | 6.3 | 0.26 |
| AJ269537 | Chondroitin-4-sulfotransferase | 60.5 | 0.26 |
| AL137555 | Is unknown | 8.5 | 0.26 |
| U89505 | RNA binding motif proteins | 5.5 | 0.26 |
| U82938 | CD27 binding proteins | 7.5 | 0.26 |
| X99584 | SMT3 homolog 1 | 12.8 | 0.26 |
| AK000847 | Is unknown | 35.8 | 0.27 |
| NM_014463 | Lsm3 protein | 7.8 | 0.27 |
| AL133645 | Is unknown | 50.8 | 0.27 |
| X78924 | Zinc finger protein 266 | 13.6 | 0.27 |
| NM_004304 | Malignant lymphoma kinase | 15.0 | 0.27 |
| X57958 | Ribosomal protein L7 | 27.9 | 0.27 |
| U63542 | Is unknown | 12.3 | 0.27 |
| AK000086 | Hypothetical proteins | 8.3 | 0.27 |
| X57138 | H2A histidine family memberMember N | 32.0 | 0.27 |
| AB023206 | KIAA0989 protein | 6.5 | 0.27 |
| AB021641 | Gonadotropin-induced transcription repressor 1 | 5.5 | 0.28 |
| AF050639 | NADH dehydrogenase | 5.5 | 0.28 |
| M62505 | Complement component 5 receptor 1 | 7.5 | 0.28 |
| X64364 | basigin | 5.8 | 0.28 |
| AJ224082 | Is unknown | 22.5 | 0.28 |
| AF042165 | Cytochrome c oxidase | 20.4 | 0.28 |
| AK001472 | anillin | 10.9 | 0.28 |
| X86428 | Protein phosphatase 2A subunit | 12.7 | 0.28 |
| AF227132 | Candidate taste receptor T2R5 | 5.1 | 0.28 |
| Z98751 | Is unknown | 5.3 | 0.28 |
| D21260 | Clathrin heavy polypeptide | 8.3 | 0.28 |
| AF041474 | Actin-like 6 | 15.1 | 0.28 |
| NM_005258 | GTP cyclohydrolase I protein | 7.6 | 0.28 |
| L20859 | Solute carrier family 20 | 9.6 | 0.29 |
| Z80783 | H2B histone family member | 9.0 | 0.29 |
| AB011105 | Laminin alpha 5 | 7.1 | 0.29 |
| AL008726 | Beta-galactosidase protective protein | 5.2 | 0.29 |
| D29012 | Proteasome subunits | 12.6 | 0.29 |
| X63629 | Cadherin 3P-cadherin | 6.8 | 0.29 |
| X02419 | Plasminogen activator urokinase | 12.9 | 0.29 |
| X13238 | Cytochrome c oxidase | 8.0 | 0.29 |
| X59798 | Cyclin D1 | 12.7 | 0.30 |
| D78151 | Proteasome 26S subunit | 7.6 | 0.31 |
| AF054185 | Proteasome subunits | 18.8 | 0.31 |
| J03890 | Associated with the lungSurfactant protein C | 5.5 | 0.32 |
| M34079 | Proteasome 26S subunit | 5.2 | 0.33 |
[00116] Table 31: upregulation of polynucleotide expression in a549 cells induced by the peptide of formula a. A peptide concentration of 50. mu.g/m 1 was found to increase the expression of many polynucleotides. The peptides were incubated with Human a549 epithelial cells for 4 hours, RNA was isolated, converted to labeled cDNA probes, and hybridized to a Human Operon array (PRHU 04). The intensity of the polynucleotide in the unstimulated control cells is shown in the third and fourth columns, which correspond to the cDNAs labeled with the dyes Cy3 and Cy5, respectively. "ID #: control "column" refers to the intensity of polynucleotide expression in cells stimulated with peptide divided by the intensity of non-stimulated cells.
| Registration number | Gene | control-Cy 3 | control-Cy 5 | ID 5: control | ID 6: control | ID 7: control | ID 8: control | ID 9: control | ID 10: control |
| U12472 | Glutathione S-transferase | 0.09 | 0.31 | 13.0 | 3.5 | 4.5 | 7.0 | 4.3 | 16.4 |
| X66403 | Cholinergic receptors | 0.17 | 0.19 | 7.8 | 9.9 | 6.0 | 6.4 | 5.0 | 15.7 |
| AK001932 | Is unknown | 0.11 | 0.25 | 19.4 | 4.6 | 9.9 | 7.6 | 8.1 | 14.5 |
| X58079 | S100 calcium binding protein | 0.14 | 0.24 | 12.2 | 7.6 | 8.1 | 4.3 | 4.5 | 13.2 |
| U18244 | Solute carrier family 1 | 0.19 | 0.20 | 6.1 | 9.7 | 11.9 | 5.0 | 3.7 | 10.6 |
| U20648 | Zinc finger proteins | 0.16 | 0.13 | 5.3 | 6.2 | 5.6 | 3.1 | 6.8 | 9.5 |
| AB037832 | Is unknown | 0.10 | 0.29 | 9.0 | 4.2 | 9.4 | 3.1 | 2.6 | 8.7 |
| AC002542 | Is unknown | 0.15 | 0.07 | 10.5 | 15.7 | 7.8 | 10.1 | 11.7 | 8.2 |
| M89796 | Transmembrane 4-domain, subfamily A | 0.15 | 0.14 | 2.6 | 6.1 | 7.6 | 3.5 | 13.3 | 8.1 |
| AF042163 | Cytochrome c oxidase | 0.09 | 0.19 | 3.9 | 3.2 | 7.6 | 6.3 | 4.9 | 7.9 |
| AL032821 | Vanin 2 | 0.41 | 0.23 | 2.5 | 5.2 | 3.2 | 2.1 | 4.0 | 7.9 |
| U25341 | Melatonin receptor 1B | 0.04 | 0.24 | 33.1 | 5.1 | 23.3 | 6.6 | 4.1 | 7.6 |
| U52219 | G protein-coupled receptors | 0.28 | 0.20 | 2.1 | 6.2 | 6.9 | 2.4 | 3.9 | 7.1 |
| X04506 | Apolipoprotein B | 0.29 | 0.32 | 7.9 | 3.4 | 3.3 | 4.8 | 2.6 | 7.0 |
| AB011138 | Type IV ATPase | 0.12 | 0.07 | 3.5 | 12.9 | 6.6 | 6.4 | 21.3 | 6.9 |
| AF055018 | Is unknown | 0.28 | 0.22 | 3.8 | 6.9 | 5.0 | 2.3 | 3.1 | 6.8 |
| AK002037 | Hypothetical proteins | 0.08 | 0.08 | 2.9 | 7.9 | 14.1 | 7.9 | 20.1 | 6.5 |
| AK001024 | Guanylic acid binding protein | 0.16 | 0.11 | 7.7 | 11.9 | 5.0 | 10.3 | 6.0 | 6.3 |
| AF240467 | TLR-7 | 0.11 | 0.10 | 20.4 | 9.0 | 3.4 | 9.4 | 12.9 | 6.1 |
| AF105367 | Glucagon-like polypeptide 2 receptor | 0.15 | 0.35 | 23.2 | 2.6 | 3.0 | 10.6 | 2.9 | 5.7 |
| AL009183 | TNFR superfamily, member 9 | 0.46 | 0.19 | 10.6 | 4.7 | 3.7 | 2.8 | 6.5 | 5.7 |
| X54380 | Pregnancy zone proteins | 0.23 | 0.08 | 4.7 | 11.9 | 7.2 | 12.7 | 3.8 | 5.5 |
| AL137736 | Is unknown | 0.22 | 0.15 | 2.1 | 7.2 | 3.3 | 7.1 | 4.6 | 5.5 |
| X05615 | Thyroglobulin | 0.28 | 0.42 | 6.3 | 2.7 | 7.7 | 2.4 | 3.1 | 5.4 |
| D28114 | Myelin-associated proteins | 0.24 | 0.08 | 2.5 | 15.9 | 13.0 | 7.1 | 13.7 | 5.4 |
| AK000358 | Microfibrillar associated protein 3 | 0.28 | 0.28 | 8.7 | 4.2 | 7.2 | 3.2 | 2.4 | 5.3 |
| AK001351 | Is unknown | 0.12 | 0.22 | 3.9 | 7.6 | 8.7 | 3.9 | 2.3 | 5.2 |
| U79289 | Is unknown | 0.14 | 0.27 | 2.5 | 2.7 | 2.8 | 2.0 | 4.3 | 5.1 |
| AB014546 | Zinc finger proteins | 0.12 | 0.34 | 6.8 | 2.4 | 4.1 | 2.7 | 2.0 | 5.0 |
| AL117428 | DKFZP434A236 protein | 0.10 | 0.07 | 2.8 | 16.1 | 12.8 | 9.7 | 14.2 | 4.9 |
| AL050378 | Is unknown | 0.41 | 0.14 | 3.5 | 8.7 | 11.7 | 3.5 | 7.0 | 4.9 |
| AJ250562 | Transmembrane 4 superfamily member 2 | 0.13 | 0.10 | 5.2 | 5.7 | 14.2 | 3.8 | 10.3 | 4.8 |
| NM_001756 | Corticosteroid hormone binding globulin | 0.28 | 0.13 | 4.0 | 7.9 | 6.5 | 14.9 | 5.6 | 4.8 |
| AL137471 | Hypothetical proteins | 0.29 | 0.05 | 3.7 | 18.0 | 6.2 | 7.2 | 16.3 | 4.7 |
| M19684 | Protease inhibitor 1 | 0.41 | 0.14 | 3.5 | 4.6 | 5.4 | 2.8 | 9.4 | 4.7 |
| NM_001963 | Epidermal growth factor | 0.57 | 0.05 | 3.4 | 6.2 | 1.8 | 32.9 | 14.7 | 4.4 |
| NM_000910 | Neuropeptide Y receptors | 0.62 | 0.36 | 3.1 | 2.7 | 2.3 | 2.6 | 3.1 | 4.4 |
| AF022212 | Rho GTP enzyme activator protein 6 | 0.19 | 0.02 | 9.0 | 45.7 | 25.6 | 12.4 | 72.2 | 4.4 |
| AK001674 | Cofactor required for Sp1 | 0.11 | 0.13 | 8.4 | 6.5 | 7.9 | 4.5 | 7.4 | 4.3 |
| U51920 | Signal recognition particles | 0.23 | 0.27 | 3.4 | 3.8 | 2.1 | 4.1 | 8.8 | 4.2 |
| AK000576 | Hypothetical proteins | 0.27 | 0.06 | 4.4 | 14.7 | 7.4 | 14.1 | 8.6 | 4.2 |
| AL080073 | Is unknown | 0.17 | 0.20 | 21.6 | 3.9 | 4.3 | 8.8 | 2.6 | 4.1 |
| U59628 | Paired cassette Gene 9 | 0.34 | 0.06 | 3.4 | 14.1 | 5.4 | 7.9 | 4.9 | 4.1 |
| U90658 | Lactophagous protein, subfamily 3, member A3 | 0.41 | 0.31 | 2.3 | 4.7 | 5.5 | 6.8 | 3.4 | 4.1 |
| M19673 | Caspase inhibitors SA | 0.43 | 0.26 | 2.3 | 8.5 | 4.5 | 2.5 | 4.1 | 3.8 |
| AL161972 | ICAM 2 | 0.44 | 0.37 | 2.0 | 3.6 | 2.0 | 2.7 | 5.5 | 3.8 |
| X54938 | Inositol 1, 4, 5-trisphosphate 3-kinase A | 0.32 | 0.22 | 3.9 | 3.3 | 6.2 | 3.1 | 4.4 | 3.7 |
| AB014575 | KIAA0675 gene product | 0.04 | 0.13 | 46.2 | 4.5 | 10.2 | 8.0 | 6.2 | 3.4 |
| M83664 | MHC II,DP β1 | 0.57 | 0.29 | 2.9 | 2.1 | 2.0 | 3.1 | 6.6 | 3.4 |
| AK000043 | Hypothetical proteins | 0.34 | 0.14 | 2.7 | 7.1 | 3.7 | 9.4 | 8.8 | 3.3 |
| U60666 | Testis-specific leucine-rich repeat protein | 0.21 | 0.11 | 9.9 | 9.0 | 4.1 | 5.5 | 13.0 | 3.3 |
| AK000337 | Hypothetical proteins | 0.49 | 0.19 | 4.3 | 5.1 | 4.7 | 10.6 | 7.1 | 3.3 |
| AF050198 | Inferred as mitochondrial space protein | 0.34 | 0.15 | 7.0 | 6.3 | 3.6 | 5.6 | 11.9 | 3.3 |
| AJ251029 | Odour binding protein 2A | 0.28 | 0.12 | 4.4 | 9.4 | 7.2 | 8.8 | 7.1 | 3.2 |
| X74142 | Forked head type frame G1B | 0.12 | 0.33 | 19.5 | 4.5 | 8.4 | 6.4 | 4.4 | 3.2 |
| AB029033 | KIAA1110 protein | 0.35 | 0.24 | 3.1 | 2.2 | 5.6 | 5.2 | 3.1 | 3.1 |
| D85606 | Cholecystokinin A receptors | 0.51 | 0.14 | 4.3 | 3.9 | 4.6 | 3.5 | 7.2 | 3.1 |
| X84195 | Acylphosphatase 2 muscle type | 0.32 | 0.19 | 4.8 | 3.7 | 5.0 | 11.2 | 9.8 | 3.0 |
| U57971 | ATP enzyme calcium ion transport cytoplasmic Membrane 3 | 0.29 | 0.13 | 2.2 | 7.9 | 1.8 | 6.3 | 4.8 | 3.0 |
| J02611 | Apolipoprotein D | 0.28 | 0.10 | 2.8 | 11.0 | 3.7 | 10.3 | 8.4 | 3.0 |
| AF071510 | Lecithin retinol acyltransferase | 0.07 | 0.05 | 7.9 | 3.8 | 11.7 | 46.0 | 16.3 | 3.0 |
| AF131757 | Is unknown | 0.10 | 0.08 | 4.8 | 9.0 | 44.3 | 9.3 | 10.7 | 3.0 |
| L10717 | IL 2-inducible T cell kinase | 0.45 | 0.21 | 2.5 | 4.9 | 2.8 | 10.9 | 4.5 | 2.9 |
| L32961 | 4-aminobutyrate aminotransferase | 0.64 | 0.32 | 3.6 | 2.9 | 3.2 | 5.3 | 2.3 | 2.9 |
| NM_003631 | Poly (ADP-ribose) glycohydrolases | 0.46 | 0.41 | 9.7 | 3.9 | 4.1 | 3.8 | 2.8 | 2.7 |
| AF098484 | pronaspin A | 0.28 | 0.14 | 3.7 | 3.7 | 5.6 | 11.6 | 3.7 | 2.5 |
| NM_009589 | Acylsulfatase D | 0.73 | 0.16 | 3.2 | 5.6 | 6.0 | 48.6 | 7.2 | 2.4 |
| M14764 | TNFR superfamily, member 16 | 0.49 | 0.15 | 2.3 | 3.5 | 10.6 | 13.6 | 6.8 | 2.2 |
| AL035250 | Endotoxin 3 | 0.52 | 0.14 | 2.1 | 7.3 | 4.8 | 4.5 | 3.7 | 2.2 |
| M97925 | Defensin, alpha 5, Paneth cell specificity | 0.33 | 0.07 | 4.0 | 14.7 | 7.8 | 9.4 | 3.5 | 2.1 |
| D43945 | Transcription factor EC | 0.46 | 0.19 | 6.6 | 2.9 | 8.2 | 4.0 | 3.5 | 2.1 |
| D16583 | Histidine decarboxylase | 0.46 | 0.09 | 3.2 | 13.8 | 4.2 | 8.8 | 13.7 | 2.1 |
[00117] Table 32: upregulation of polynucleotide expression in a549 cells induced by a peptide of formula B. A peptide concentration of 50. mu.g/ml was found to increase the expression of many polynucleotides. The peptides were incubated with human a549 epithelial cells for 4 hours, RNA was isolated, converted to labeled cDNA probes, and hybridized to a human operon array (PRHU 04). The intensity of the polynucleotide in the unstimulated control cells is shown in the third and fourth columns, which correspond to the cDNAs labeled with the dyes Cy3 and Cy5, respectively. "ID #: control "column" refers to the intensity of polynucleotide expression in cells stimulated with peptide divided by the intensity of non-stimulated cells.
| Registration number | Gene | control-Cy 3 | control-Cy 5 | ID 12: control | ID 13: control | ID 14: control | ID 15: control | ID 16: control | ID 17: control |
| AL157466 | Is unknown | 0.05 | 0.06 | 18.0 | 21.4 | 16.7 | 5.2 | 6.8 | 8.6 |
| AB023215 | KIAA0998 protein | 0.19 | 0.07 | 14.8 | 10.6 | 7.9 | 14.4 | 6.6 | 16.1 |
| AL031121 | Is unknown | 0.24 | 0.08 | 14.1 | 5.7 | 3.8 | 5.5 | 2.8 | 4.6 |
| NM_016331 | Zinc finger proteins | 0.16 | 0.08 | 12.8 | 7.2 | 11.0 | 5.3 | 11.2 | 9.7 |
| M14565 | Cytochrome P450 | 0.16 | 0.12 | 10.6 | 12.5 | 5.0 | 3.6 | 10.1 | 6.3 |
| U22492 | G protein-coupled receptors | 0.28 | 0.07 | 10.4 | 8.9 | 4.8 | 10.8 | 6.6 | 3.6 |
| U76010 | Solute carrier family 30 | 0.14 | 0.07 | 9.7 | 18.6 | 3.7 | 4.8 | 5.6 | 8.9 |
| AK000685 | Is unknown | 0.51 | 0.10 | 9.0 | 3.1 | 2.8 | 3.9 | 15.3 | 3.0 |
| AF013620 | Immunoglobulin heavy chain variable region 4-4 | 0.19 | 0.18 | 8.5 | 2.6 | 6.2 | 5.7 | 8.2 | 3.8 |
| AL049296 | Is unknown | 0.61 | 0.89 | 8.1 | 3.2 | 2.7 | 3.2 | 2.7 | 2.0 |
| AB006622 | KIAA0284 protein | 0.47 | 0.28 | 7.5 | 5.0 | 2.8 | 11.1 | 5.5 | 4.6 |
| X04391 | CD5 antigen | 0.22 | 0.13 | 7.2 | 16.7 | 2.7 | 7.7 | 6.1 | 5.9 |
| AK000067 | Hypothetical proteins | 0.80 | 0.35 | 7.1 | 4.6 | 2.1 | 3.2 | 8.5 | 2.2 |
| AF053712 | TNF superfamily member 11 | 0.17 | 0.08 | 6.9 | 17.7 | 3.0 | 6.2 | 12.3 | 5.2 |
| X58079 | S100 calcium binding protein A1 | 0.14 | 0.24 | 6.7 | 6.7 | 5.9 | 6.5 | 5.3 | 2.5 |
| M91036 | Hemoglobin _ Gamma A | 0.48 | 0.36 | 6.7 | 14.2 | 2.1 | 2.9 | 2.7 | 4.8 |
| AF055018 | Is unknown | 0.28 | 0.22 | 6.3 | 10.7 | 2.7 | 2.6 | 4.6 | 6.5 |
| L17325 | Pre-T/NK cell related proteins | 0.19 | 0.29 | 6.1 | 4.4 | 6.5 | 4.7 | 4.0 | 4.0 |
| D45399 | Phosphodiesterase enzyme | 0.21 | 0.18 | 6.1 | 4.6 | 5.0 | 2.8 | 10.8 | 4.0 |
| AB023188 | KIAA0971 protein | 0.29 | 0.13 | 5.9 | 10.6 | 3.6 | 3.4 | 10.6 | 7.2 |
| NM_012177 | F-box protein | 0.26 | 0.31 | 5.9 | 5.5 | 3.8 | 2.8 | 3.0 | 6.8 |
| D38550 | E2F TF3 | 0.43 | 0.39 | 5.8 | 3.4 | 2.1 | 4.5 | 2.5 | 2.4 |
| AL050219 | Is unknown | 0.26 | 0.04 | 5.7 | 17.0 | 3.1 | 9.2 | 30.3 | 16.1 |
| AL137540 | Is unknown | 0.67 | 0.79 | 5.5 | 3.2 | 3.9 | 10.9 | 2.9 | 2.3 |
| D50926 | KIAA0136 protein | 0.57 | 0.21 | 5.4 | 5.6 | 2.0 | 3.3 | 4.4 | 3.2 |
| AL137658 | Is unknown | 0.31 | 0.07 | 5.4 | 12.1 | 2.6 | 10.8 | 3.9 | 8.6 |
| U21931 | Fructose bisphosphatase 1 | 0.48 | 0.14 | 5.4 | 4.1 | 2.9 | 3.6 | 6.0 | 3.2 |
| AK001230 | DKFZP586D211 protein | 0.43 | 0.26 | 5.0 | 4.6 | 2.1 | 2.2 | 2.5 | 2.7 |
| AL137728 | Is unknown | 0.67 | 0.47 | 5.0 | 5.9 | 2.2 | 6.8 | 5.9 | 2.1 |
| AB022847 | Is unknown | 0.39 | 0.24 | 4.5 | 2.2 | 3.5 | 4.3 | 3.8 | 3.7 |
| X75311 | Mevalonate kinase | 0.67 | 0.22 | 4.3 | 4.0 | 2.0 | 8.3 | 4.0 | 5.1 |
| AK000946 | DKFZP566C243 protein | 0.36 | 0.29 | 4.1 | 3.8 | 3.9 | 5.4 | 25.8 | 2.7 |
| AB023197 | KIAA0980 protein | 0.25 | 0.30 | 4.0 | 8.3 | 2.1 | 8.8 | 2.2 | 4.9 |
| AB014615 | Fibroblast growth factor 8 | 0.19 | 0.07 | 3.9 | 3.3 | 7.0 | 3.4 | 2.2 | 7.7 |
| X04014 | Is unknown | 0.29 | 0.16 | 3.8 | 2.5 | 2.2 | 3.0 | 5.5 | 3.1 |
| U76368 | Solute carrier family 7 | 0.46 | 0.17 | 3.8 | 3.8 | 2.8 | 3.2 | 4.2 | 3.0 |
| AB032436 | Is unknown | 0.14 | 0.21 | 3.8 | 2.7 | 6.1 | 3.2 | 4.5 | 2.6 |
| AB020683 | KIAA0876 protein | 0.37 | 0.21 | 3.7 | 4.2 | 2.2 | 5.3 | 2.9 | 9.4 |
| NM_012126 | Saccharide sulfotransferase 5 | 0.31 | 0.20 | 3.7 | 5.2 | 3.2 | 3.4 | 3.9 | 2.5 |
| AK002037 | Hypothetical proteins | 0.08 | 0.08 | 3.7 | 17.1 | 4.6 | 12.3 | 11.0 | 8.7 |
| X78712 | Glycerol kinase pseudogene 2 | 0.17 | 0.19 | 3.6 | 2.5 | 4.5 | 5.3 | 2.2 | 3.3 |
| NM_014178 | HSPC156 protein | 0.23 | 0.12 | 3.5 | 8.4 | 2.9 | 6.9 | 14.4 | 5.5 |
| AC004079 | Homology box A2 | 0.31 | 0.11 | 3.5 | 7.0 | 2.1 | 2.0 | 7.3 | .9.1 |
| AL080182 | Is unknown | 0.51 | 0.21 | 3.4 | 3.5 | 2.2 | 2.1 | 2.9 | 2.4 |
| M91036 | Hemoglobin gamma G | 0.22 | 0.02 | 3.4 | 26.3 | 5.8 | 6.8 | 30.4 | 21.6 |
| AJ000512 | Serum/glucocorticoid-regulated kinase | 0.27 | 0.43 | 3.3 | 2.1 | 4.9 | 2.3 | 3.9 | 2.7 |
| AK002140 | Hypothetical proteins | 0.28 | 0.14 | 3.3 | 9.9 | 2.8 | 2.1 | 16.6 | 7.2 |
| AL137284 | Is unknown | 0.22 | 0.04 | 3.3 | 7.2 | 4.1 | 6.0 | 12.2 | 3.7 |
| Z11898 | POU Domain _ type 5TF1 | 0.12 | 0.29 | 3.2 | 3.7 | 8.2 | 2.5 | 6.6 | 2.2 |
| AB017016 | Brain specific proteins | 0.27 | 0.29 | 3.1 | 2.8 | 2.5 | 2.8 | 3.3 | 5.5 |
| X54673 | Solute carrier family 6 | 0.34 | 0.08 | 2.9 | 12.0 | 2.2 | 10.4 | 7.4 | 5.9 |
| AL033377 | Is unknown | 0.40 | 0.22 | 2.6 | 2.6 | 2.6 | 2.3 | 4.5 | 2.2 |
| X85740 | CCR4 | 0.34 | 0.05 | 2.6 | 2.3 | 2.6 | 2.5 | 12.5 | 5.2 |
| AB010419 | Core binding proteins | 0.59 | 0.20 | 2.5 | 12.8 | 2.0 | 2.8 | 2.9 | 5.9 |
| AL109726 | Is unknown | 0.14 | 0.15 | 2.3 | 9.0 | 4.3 | 4.4 | 2.6 | 3.7 |
| NM_012450 | Sulfate Transporter 1 | 0.15 | 0.10 | 2.2 | 3.1 | 8.2 | 9.9 | 4.7 | 5.9 |
| J04599 | Biglycan proteoglycan | 0.39 | 0.30 | 2.1 | 3.3 | 6.6 | 2.2 | 2.7 | 5.4 |
| AK000266 | Hypothetical proteins | 0.49 | 0.35 | 2.1 | 3.5 | 3.5 | 6.6 | 4.3 | 4.0 |
[00118] Table 33: upregulation of polynucleotide expression in a549 cells induced by the peptide of formula C. A peptide concentration of 50. mu.g/ml was found to increase the expression of many polynucleotides. The peptides were incubated with human a549 epithelial cells for 4 hours, RNA was isolated, converted to labeled cDNA probes, and hybridized to a human operon array (PRHU 04). The intensity of the polynucleotide in the unstimulated control cells is shown in the third and fourth columns, which correspond to the cDNAs labeled with the dyes Cy3 and Cy5, respectively. "ID #: control "column" refers to the intensity of polynucleotide expression in cells stimulated with peptide divided by the intensity of non-stimulated cells.
| Registration number | Gene | control-Cy 3 | control-Cy 5 | ID 19: control | ID20: control | ID 21: control | ID 22: control | ID 23: control | ID 24: control |
| NM_014139 | Voltage-gated sodium channels | 0.04 | 0.05 | 31.6 | 25.2 | 18.0 | 9.7 | 22.2 | 11.2 |
| X84003 | TATA box binding proteins | 0.47 | 0.07 | 31.8 | 12.7 | 2.5 | 2.8 | 18.0 | 14.2 |
| AF144412 | Lens epithelial cell proteins | 0.25 | 0.07 | 23.9 | 8.0 | 6.8 | 3.4 | 16.2 | 3.5 |
| AL080107 | Is unknown | 0.11 | 0.06 | 17.8 | 34.4 | 12.4 | 6.2 | 5.4 | 7.9 |
| AF052116 | Is unknown | 0.34 | 0.07 | 15.5 | 3.9 | 9.2 | 3.0 | 6.9 | 2.7 |
| AB033063 | Is unknown | 0.46 | 0.13 | 15.2 | 10.3 | 4.0 | 2.6 | 7.2 | 11.2 |
| AK000258 | Hypothetical proteins | 0.27 | 0.07 | 13.9 | 8.0 | 3.5 | 3.4 | 26.5 | 11.5 |
| NM_006963 | Zinc finger proteins | 0.10 | 0.08 | 12.8 | 6.8 | 6.2 | 5.9 | 17.2 | 1241.2 |
| NM_014099 | PRO1768 protein | 0.30 | 0.06 | 12.3 | 17.4 | 5.4 | 5.4 | 19.5 | 3.4 |
| AK000996 | Hypothetical proteins | 0.17 | 0.07 | 10.0 | 8.0 | 9.7 | 7.4 | 20.7 | 16.3 |
| M81933 | Cell division cyclin 25A | 0.13 | 0.21 | 8.8 | 7.8 | 19.6 | 15.6 | 4.8 | 3.8 |
| AF181286 | Is unknown | 0.05 | 0.22 | 8.8 | 2.7 | 12.0 | 35.6 | 5.9 | 2.3 |
| AJ272208 | IL-1R helper protein-like 2 | 0.22 | 0.17 | 8.8 | 2.9 | 5.0 | 3.2 | 9.8 | 7.3 |
| AF030555 | Fatty acid coenzyme A ligase | 0.10 | 0.39 | 8.7 | 2.2 | 11.3 | 9.9 | 3.0 | 2.1 |
| AL050125 | Is unknown | 0.23 | 0.07 | 8.6 | 14.3 | 5.2 | 2.8 | 18.7 | 8.3 |
| AB011096 | KIAA0524 protein | 0.21 | 0.08 | 8.5 | 24.4 | 4.7 | 6.8 | 10.4 | 7.5 |
| J03068 | N-acylaminoacyl-peptide hydrolases | 0.54 | 0.21 | 8.3 | 2.4 | 2.2 | 4.1 | 3.0 | 6.0 |
| M33906 | Class II MHC, DQ α 1 | 0.14 | 0.08 | 7.6 | 4.5 | 15.2 | 6.1 | 7.5 | 7.9 |
| AJ272265 | Secretion of phosphoproteins | 0.21 | 0.09 | 7.6 | 9.0 | 3.3 | 4.9 | 18.8 | 14.5 |
| J00210 | Interferon alpha 13 | 0.41 | 0.07 | 7.2 | 15.0 | 2.8 | 3.1 | 11.0 | 4.3 |
| AK001952 | Hypothetical proteins | 0.42 | 0.21 | 6.9 | 4.9 | 2.5 | 3.1 | 7.6 | 4.5 |
| X54131 | Protein tyrosine phosphatases, receptor types | 0.09 | 0.20 | 6.4 | 6.5 | 7.7 | 15.0 | 5.6 | 4.1 |
| AF064493 | LIM binding Domain 2 | 0.46 | 0.14 | 5.9 | 5.6 | 2.2 | 2.9 | 8.5 | 5.8 |
| AL117567 | DKFZP566O084 protein | 0.44 | 0.22 | 5.8 | 3.3 | 2.9 | 2.3 | 5.7 | 14.9 |
| L40933 | Phosphoglucomutase 5 | 0.16 | 0.03 | 5.6 | 11.0 | 4.8 | 3.5 | 8.5 | 76.3 |
| M27190 | regenerating islet-derived 1 alpha (pancreatic stone protein).) | 0.19 | 0.28 | 5.3 | 3.0 | 3.8 | 3.6 | 5.8 | 3.6 |
| AL031121 | Is unknown | 0.24 | 0.09 | 5.3 | 3.8 | 3.2 | 3.9 | 3.0 | 27.9 |
| U27655 | Modulators of G protein signaling | 0.24 | 0.29 | 5.0 | 9.0 | 4.5 | 8.3 | 4.2 | 4.5 |
| AB037786 | Is unknown | 0.12 | 0.03 | 4.7 | 54.1 | 2.8 | 2.3 | 2.2 | 11.0 |
| X73113 | Myosin binding protein C | 0.29 | 0.13 | 4.7 | 6.5 | 6.0 | 2.4 | 6.7 | 6.3 |
| AB010962 | Matrix metalloproteinases | 0.08 | 0.12 | 4.7 | 6.2 | 2.4 | 4.7 | 10.9 | 4.2 |
| AL096729 | Is unknown | 0.36 | 0.13 | 4.7 | 7.7 | 3.2 | 2.4 | 6.3 | 6.2 |
| AB018320 | Arg/Ab1 interacting proteins | 0.16 | 0.18 | 4.6 | 7.1 | 3.0 | 3.3 | 5.8 | 8.9 |
| AK001024 | Guanylic acid binding protein | 0.16 | 0.11 | 4.6 | 2.0 | 9.8 | 2.6 | 7.6 | 14.1 |
| AJ21931 | Is unknown | 0.15 | 0.08 | 4.6 | 17.3 | 5.4 | 9.2 | 5.1 | 5.5 |
| U21931 | Fructose bisphosphatase 1 | 0.48 | 0.14 | 4.6 | 4.3 | 2.6 | 2.1 | 8.4 | 9.6 |
| X66403 | Cholinergic receptors | 0.17 | 0.19 | 4.4 | 9.0 | 10.9 | 9.3 | 5.1 | 6.7 |
| X67734 | contactin 2 | 0.25 | 0.09 | 4.3 | 6.8 | 3.1 | 5.8 | 7.9 | 8.4 |
| U92981 | Is unknown | 0.20 | 0.23 | 4.3 | 3.2 | 4.8 | 5.6 | 5.4 | 6.3 |
| X68879 | Air door homolog 1 | 0.05 | 0.08 | 4.3 | 2.0 | 12.3 | 2.7 | 5.6 | 4.7 |
| AL137362 | Is unknown | 0.22 | 0.22 | 4.2 | 4.1 | 2.7 | 4.1 | 9.3 | 4.2 |
| NM_001756 | Corticosteroid hormone binding globulin | 0.28 | 0.13 | 4.4 | 10.6 | 3.9 | 2.7 | 10.3 | 5.5 |
| U80770 | Is unknown | 0.31 | 0.14 | 4.1 | 4.1 | 23.3 | 2.7 | 7.0 | 10.1 |
| AL109792 | Is unknown | 0.16 | 0.19 | 4.0 | 4.5 | 4.3 | 8.8 | 8.7 | 3.9 |
| X65962 | Cytosome rope P450 | 0.33 | 0.05 | 3.8 | 25.3 | 5.7 | 5.1 | 19.8 | 12.0 |
| AK001856 | Is unknown | 0.40 | 0.21 | 3.8 | 7.0 | 2.6 | 3.1 | 2.9 | 7.8 |
| AL022723 | MHC, class I, F | 0.55 | 0.18 | 3.7 | 5.7 | 4.4 | 2.3 | 3.3 | 5.2 |
| D38449 | Putative G protein-coupled receptors | 0.18 | 0.09 | 3.5 | 11.1 | 13.3 | 5.8 | 4.8 | 5.2 |
| AL137489 | Is unknown | 0.74 | 0.26 | 3.3 | 2.9 | 2.6 | 3.3 | 2.5 | 5.4 |
| AB000887 | Small molecule inducible cytokine subfamily A | 0.76 | 0.18 | 3.3 | 5.0 | 2.6 | 2.4 | 5.9 | 10.3 |
| NM_012450 | Sulfate Transporter 1 | 0.15 | 0.10 | 3.3 | 9.0 | 10.0 | 10.9 | 4.6 | 8.7 |
| U86529 | Glutathione S-transferase ζ 1 | 0.55 | 0.15 | 3.2 | 6.8 | 4.4 | 2.3 | 9.3 | 5.1 |
| AK001244 | Is unknown | 0.79 | 0.31 | 3.2 | 5.5 | 2.3 | 2.3 | 3.9 | 2.8 |
| AL133602 | Is unknown | 0.16 | 0.21 | 3.1 | 7.8 | 8.7 | 2.6 | 4.1 | 5.6 |
| AB033080 | Cell cycle process 8 protein | 0.31 | 0.31 | 3.1 | 4.6 | 3.0 | 3.5 | 2.2 | 4.2 |
| AF023466 | Putative glycine-N-acyltransferase | 0.27 | 0.18 | 3.1 | 5.0 | 4.2 | 7.4 | 10.1 | 3.8 |
| AL117457 | Actin (cofilin)2 | 0.68 | 0.53 | 3.0 | 4.6 | 3.3 | 2.4 | 7.4 | 3.4 |
| AC007059 | Is unknown | 0.37 | 0.35 | 3.0 | 5.7 | 3.1 | 2.4 | 2.6 | 2.4 |
| U60179 | Growth hormone receptors | 0.34 | 0.21 | 2.9 | 3.5 | 2.3 | 3.1 | 8.0 | 4.7 |
| M37238 | Phospholipase C, gamma 2 | 0.60 | 0.36 | 2.9 | 2.0 | 3.2 | 2.1 | 2.9 | 4.6 |
| L22569 | Cathepsin B | 0.32 | 0.12 | 2.9 | 2.1 | 6.2 | 3.0 | 13.1 | 16.7 |
| M80359 | MAP/microtubule affinity regulated kinase 3 | 0.37 | 0.76 | 2.9 | 3.1 | 6.1 | 7.6 | 2.1 | 3.3 |
| S70348 | Integrin beta 3 | 0.58 | 0.31 | 2.6 | 4.8 | 4.1 | 2.6 | 2.6 | 2.6 |
| L13720 | Growth retardation specific protein 6 | 0.36 | 0.26 | 2.4 | 2.5 | 6.8 | 4.8 | 3.9 | 3.7 |
| AL049423 | Is unknown | 0.33 | 0.30 | 2.4 | 3.7 | 3.8 | 2.8 | 2.9 | 3.4 |
| AL050201 | Is unknown | 0.68 | 0.29 | 2.2 | 3.1 | 3.7 | 3.0 | 3.0 | 2.2 |
| AF050078 | Growth ofBlocking specific protein 11 | 0.87 | 0.33 | 2.1 | 8.4 | 2.5 | 2.2 | 2.6 | 4.4 |
| AK001753 | Hypothetical proteins | 0.53 | 0.28 | 2.1 | 5.0 | 2.2 | 2.8 | 3.6 | 4.6 |
| X05323 | Is unknown | 0.39 | 0.13 | 2.1 | 7.8 | 2.6 | 2.4 | 21.5 | 3.5 |
| AB014548 | KIAA0648 protein | 0.61 | 0.30 | 2.0 | 2.4 | 4.8 | 3.4 | 4.9 | 3.9 |
[00119] Table 34: upregulation of polynucleotide expression in a549 cells induced by the peptide of formula D. A peptide concentration of 50. mu.g/ml was found to increase the expression of many polynucleotides. The peptides were incubated with human a549 epithelial cells for 4 hours, RNA was isolated, converted to labeled cDNA probes, and hybridized to a human operon array (PRHU 04). The intensity of the polynucleotide in the unstimulated control cells is shown in the third and fourth columns, which correspond to the cDNAs labeled with the dyes Cy3 and Cy5, respectively. "ID #: control "column" refers to the intensity of polynucleotide expression in cells stimulated with peptide divided by the intensity of non-stimulated cells.
| Registration number | Gene | control-Cy 3 | control-Cy 5 | ID 26: control | ID 27: control | ID 28: control | ID 29: control | ID 30: control | ID 31: control |
| U68018 | MAD homolog 2 | 0.13 | 0.71 | 11.2 | 2.2 | 8.0 | 2.3 | 6.7 | 25.6 |
| NM_016015 | CGI-68 protein | 0.92 | 1.59 | 2.3 | 2.3 | 3.5 | 3.7 | 3.4 | 22.9 |
| AF071510 | Lecithin retinol acyltransferase e | 0.07 | 0.05 | 15.4 | 10.3 | 5.3 | 44.1 | 2.1 | 21.2 |
| AC005154 | Is unknown | 0.17 | 1.13 | 2.7 | 7.2 | 12.6 | 6.4 | 3.3 | 20.6 |
| M81933 | Cell division cycle 25A | 0.13 | 0.21 | 4.3 | 3.1 | 3.2 | 4.3 | 5.6 | 18.2 |
| AF124735 | LIM frame gene 2 | 0.17 | 0.21 | 2.1 | 4.4 | 5.9 | 5.2 | 7.6 | 17.0 |
| AL110125 | Is unknown | 0.30 | 0.08 | 5.0 | 2.7 | 6.8 | 10.2 | 2.8 | 12.0 |
| NM_004732 | Voltage gated potassium channels | 0.15 | 0.16 | 7.6 | 4.0 | 3.4 | 2.2 | 2.9 | 11.4 |
| AF030555 | Fatty acid coenzyme A ligase Long chain 4 | 0.10 | 0.39 | 10.5 | 2.2 | 6.4 | 3.0 | 5.1 | 10.7 |
| AF000237 | 1-acylglycerol-3-phosphate O-acyltransferase e2 | 1.80 | 2.37 | 3.4 | 2.5 | 2.4 | 2.1 | 3.7 | 9.9 |
| AL031588 | Hypothetical proteins | 0.40 | 0.26 | 5.8 | 20.2 | 2.8 | 4.7 | 5.6 | 9.1 |
| AL080077 | Is unknown | 0.15 | 0.21 | 2.4 | 2.0 | 11.9 | 3.8 | 2.3 | 8.7 |
| NM_014366 | Presumed to be nucleotide binding protein estradiol inducibility | 0.90 | 2.52 | 2.4 | 4.3 | 2.4 | 2.6 | 3.0 | 8.6 |
| AB002359 | Phosphoribosyl formyl glycinamidine synthetase | 0.81 | 2.12 | 3.2 | 2.7 | 5.5 | 2.5 | 2.8 | 6.9 |
| U33547 | MHC class II antigen HLA DRB6 mRNA | 0.14 | 0.16 | 2.5 | 5.3 | 4.5 | 5.0 | 3.1 | 6.6 |
| AL133051 | Is unknown | 0.09 | 0.07 | 7.7 | 6.3 | 5.4 | 23.1 | 5.4 | 6.5 |
| AK000576 | Hypothetical proteins | 0.27 | 0.06 | 7.1 | 9.3 | 5.0 | 6.9 | 2.9 | 6.2 |
| AF042378 | Spindle polar body protein | 0.36 | 0.39 | 3.3 | 3.0 | 9.5 | 4.5 | 3.4 | 6.2 |
| AF093265 | Homer neuron immediate early gene 3 | 0.67 | 0.53 | 2.7 | 13.3 | 6.5 | 5.0 | 2.9 | 6.2 |
| D80000 | Mitotic chromosome segregation 1 | 1.01 | 1.56 | 3.6 | 2.5 | 4.9 | 3.2 | 6.3 | 6.1 |
| AF035309 | Proteasome 26S subunit ATPase 5 | 3.61 | 4.71 | 2.7 | 6.6 | 5.2 | 4.9 | 2.7 | 6.0 |
| M34175 | Connexin-related protein complex 2 beta 1 subunit | 4.57 | 5.13 | 3.2 | 3.1 | 4.0 | 4.6 | 2.7 | 6.0 |
| AB020659 | KIAA0852 protein | 0.18 | 0.37 | 4.1 | 7.6 | 5.7 | 4.8 | 2.5 | 5.7 |
| NM_004862 | TNF alpha factor induced by LPS | 2.61 | 3.36 | 3.8 | 4.8 | 4.1 | 4.9 | 3.2 | 5.6 |
| U00115 | Zinc finger protein 51 | 0.51 | 0.07 | 18.9 | 2.2 | 3.5 | 7.2 | 21.2 | 5.6 |
| AF088868 | fibrousheathin II | 0.45 | 0.20 | 4.7 | 10.0 | 3.2 | 6.4 | 6.0 | 5.6 |
| AK001890 | Is unknown | 0.42 | 0.55 | 2.4 | 3.5 | 3.6 | 2.3 | 2.2 | 5.6 |
| AL137268 | KIAA0759 protein | 0.49 | 0.34 | 3.8 | 2.3 | 5.0 | 3.5 | 3.3 | 5.4 |
| X63563 | Polymerase II polypeptide B | 1.25 | 1.68 | 2.5 | 8.1 | 3.4 | 4.8 | 5.2 | 5.4 |
| D12676 | CD36 antigen | 0.35 | 0.39 | 2.9 | 3.4 | 2.6 | 2.2 | 3.5 | 5.3 |
| AK000161 | Hypothetical proteins | 1.06 | 0.55 | 3.4 | 8.7 | 2.1 | 6.7 | 2.9 | 5.1 |
| AF052138 | Is unknown | 0.64 | 0.51 | 2.9 | 2.8 | 2.7 | 5.2 | 3.6 | 5.0 |
| AL096803 | Is unknown | 0.36 | 0.03 | 20.1 | 18.3 | 3.7 | 19.3 | 16.1 | 4.9 |
| S49953 | DNA binding transcriptional activator | 0.70 | 0.15 | 3.7 | 4.0 | 2.1 | 6.6 | 4.0 | 4.8 |
| X89399 | RAS p21 protein activator | 0.25 | 0.10 | 8.5 | 14.9 | 4.8 | 18.6 | 4.3 | 4.8 |
| AJ005273 | Antigenic determinants of recA protein | 0.70 | 0.10 | 7.6 | 11.1 | 2.8 | 9.9 | 12.0 | 4.6 |
| AK001154 | Hypothetical proteins | 1.70 | 0.96 | 2.4 | 4.4 | 2.9 | 8.9 | 2.4 | 4.5 |
| AL133605 | Is unknown | 0.26 | 0.15 | 12.4 | 4.2 | 4.4 | 3.3 | 3.3 | 4.1 |
| U71092 | G protein-coupled receptor 24 | 0.53 | 0.06 | 19.0 | 9.1 | 2.2 | 12.0 | 3.3 | 4.1 |
| AF074723 | RNA polymer II transcription regulatory medium | 0.67 | 0.54 | 4.0 | 3.2 | 3.1 | 3.4 | 6.0 | 4.0 |
| AL137577 | Is unknown | 0.32 | 0.12 | 31.4 | 6.2 | 5.3 | 10.1 | 25.3 | 3.9 |
| AF151043 | Hypothetical proteins | 0.48 | 0.35 | 2.6 | 2.2 | 2.0 | 3.3 | 2.2 | 3.8 |
| AF131831 | Is unknown | 0.67 | 0.81 | 2.1 | 7.0 | 3.5 | 3.2 | 3.9 | 3.7 |
| D50405 | Histidine deacetylase 1 | 1.52 | 1.62 | 3.1 | 7.2 | 2.9 | 4.1 | 2.8 | 3.7 |
| U78305 | Protein phosphatase 1D | 1.21 | 0.20 | 4.7 | 13.0 | 3.5 | 5.9 | 4.2 | 3.7 |
| AL035562 | Paired box gene 1 | 0.24 | 0.01 | 30.2 | 81.9 | 5.6 | 82.3 | 6.2 | 3.7 |
| U67156 | Mitogen-activated protein kinase 5 | 1.15 | 0.30 | 6.6 | 3.0 | 2.2 | 2.3 | 2.5 | 3.6 |
| AL031121 | Is unknown | 0.24 | 0.09 | 5.2 | 3.7 | 2.3 | 6.5 | 9.1 | 3.6 |
| U13666 | G protein-coupled receptor 1 | 0.34 | 0.14 | 3.8 | 5.4 | 3.1 | 3.3 | 2.8 | 3.6 |
| AB018285 | KIAA0742 protein | 0.53 | 0.13 | 14.9 | 13.9 | 5.9 | 18.5 | 15.2 | 3.5 |
| D42053 | Site 1 protease | 0.63 | 0.40 | 2.6 | 7.1 | 5.6 | 9.2 | 2.6 | 3.5 |
| AK001135 | Sec23 interacting protein p125 | 0.29 | 0.53 | 5.7 | 4.5 | 3.4 | 2.6 | 11.3 | 3.4 |
| AL137461 | Is unknown | 0.25 | 0.02 | 23.8 | 9.0 | 2.7 | 59.2 | 12.5 | 3.3 |
| NM_006963 | Zinc finger protein 22 | 0.10 | 0.08 | 3.2 | 7.6 | 3.7 | 7.9 | 11.2 | 3.2 |
| AL137540 | Is unknown | 0.67 | 0.79 | 3.9 | 2.6 | 5.6 | 4.2 | 3.5 | 3.1 |
| AL137718 | Is unknown | 0.95 | 0.18 | 4.7 | 8.0 | 4.0 | 13.3 | 3.0 | 3.1 |
| AF012086 | RNA binding protein 2-like 1 | 1.20 | 0.59 | 4.6 | 4.0 | 2.0 | 4.6 | 3.6 | 3.1 |
| S57296 | HER2/neu receptor | 0.59 | 0.17 | 7.3 | 12.1 | 2.3 | 20.0 | 22.2 | 3.0 |
| NM_013329 | GC sequence-rich DNA binding factor candidate | 0.16 | 0.08 | 6.9 | 14.3 | 9.7 | 3.3 | 7.2 | 3.0 |
| AF038664 | UDP-Gal: beta GlcN Ac beta 1_ 4-galactosyltransferase | 0.15 | 0.03 | 13.4 | 22.2 | 5.4 | 15.8 | 17.6 | 3.0 |
| AF080579 | Human integral membrane protein | 0.34 | 1.03 | 3.3 | 3.0 | 6.7 | 2.1 | 2.9 | 2.9 |
| AK001075 | Hypothetical proteins | 0.67 | 0.10 | 2.1 | 2.6 | 2.6 | 8.9 | 2.2 | 2.9 |
| AB011124 | KIAA0552 Gene products | 0.46 | 0.04 | 9.6 | 72.0 | 6.0 | 33.9 | 13.6 | 2.9 |
| J03068 | N-acylaminoacyl-peptide hydrolases | 0.54 | 0.21 | 2.2 | 5.0 | 2.4 | 5.2 | 3.6 | 2.8 |
| D87120 | Osteoblast proteins | 0.87 | 0.87 | 2.2 | 2.0 | 4.7 | 2.3 | 2.0 | 2.8 |
| AB006537 | IL-1R accessory proteins | 0.17 | 0.07 | 2.9 | 7.0 | 14.5 | 5.3 | 6.6 | 2.8 |
| L34587 | Transcriptional elongation factor B | 2.49 | 1.23 | 2.2 | 16.3 | 5.0 | 15.8 | 5.5 | 2.7 |
| D31891 | SET Domain _ fork _1 | 1.02 | 0.29 | 3.9 | 6.0 | 4.3 | 4.9 | 6.6 | 2.7 |
| D00760 | Proteasome subunit alpha type 2 | 4.97 | 4.94 | 4.1 | 2.6 | 2.0 | 2.8 | 2.7 | 2.7 |
| AC004774 | digital-less homology box 5 | 0.25 | 0.12 | 2.3 | 6.3 | 3.8 | 5.2 | 5.2 | 2.6 |
| AL024493 | Is unknown | 1.46 | 0.54 | 4.8 | 13.5 | 2.1 | 11.6 | 6.8 | 2.6 |
| AB014536 | copine III | 1.80 | 1.29 | 3.2 | 9.5 | 3.8 | 6.8 | 2.6 | 2.6 |
| X59770 | IL-1R type II | 0.59 | 0.16 | 9.6 | 4.7 | 3.9 | 3.2 | 4.9 | 2.5 |
| AF052183 | Is unknown | 0.65 | 0.76 | 4.0 | 3.7 | 2.3 | 5.0 | 3.0 | 2.5 |
| AK000541 | Hypothetical proteins | 0.92 | 0.27 | 4.5 | 13.9 | 3.6 | 18.1 | 4.3 | 2.5 |
| U88528 | cAMP response element binding proteins | 1.37 | 0.86 | 3.1 | 5.4 | 2.1 | 2.8 | 2.1 | 2.4 |
| M97925 | Defensin alpha 5 paneth cell specificity | 0.33 | 0.07 | 4.6 | 35.9 | 2.0 | 7.8 | 6.5 | 2.4 |
| NM_013393 | Cell division protein FtsJ | 1.38 | 0.94 | 3.1 | 5.8 | 2.1 | 4.2 | 2.6 | 2.3 |
| X62744 | Class II MHC DM alpha | 0.86 | 0.32 | 4.0 | 4.7 | 2.3 | 2.9 | 6.1 | 2.3 |
| AF251040 | Presumed to be a nuclear protein | 0.64 | 0.30 | 6.7 | 3.4 | 2.9 | 3.9 | 5.7 | 2.2 |
| AK000227 | Hypothetical proteins | 1.49 | 0.43 | 3.4 | 7.1 | 2.3 | 3.3 | 9.1 | 2.1 |
| U88666 | SFRS protein kinase 2 | 1.78 | 0.37 | 3.4 | 5.9 | 2.6 | 8.4 | 6.1 | 2.0 |
[00120] Table 35: upregulation of polynucleotide expression in a549 cells induced by a peptide of formula E. A peptide concentration of 50. mu.g/ml was found to increase the expression of many polynucleotides. The peptides were incubated with human a549 epithelial cells for 4 hours, RNA was isolated, converted to labeled cDNA probes, and hybridized to a human operon array (PRHU 04). The intensity of the polynucleotide in the unstimulated control cells is shown in the third and fourth columns, which correspond to the cDNAs labeled with the dyes Cy3 and Cy5, respectively. "ID #: control "column" refers to the intensity of polynucleotide expression in cells stimulated with peptide divided by the intensity of non-stimulated cells.
| Registration number | Gene | control-Cy 3 | control-Cy 5 | ID 33: control | ID 34: control | ID 35: control | ID 36: control | ID 37: control | ID 38: control |
| AL049689 | Novel human mRNA | 0.25 | 0.05 | 2.7 | 26.5 | 3.3 | 21.7 | 5.4 | 37.9 |
| AK000576 | Hypothetical proteins | 0.27 | 0.06 | 3.0 | 19.1 | 3.9 | 23.0 | 3.1 | 28.3 |
| X74837 | Mannosidase, a class 1A member1 | 0.10 | 0.07 | 5.6 | 10.0 | 10.8 | 12.3 | 12.0 | 19.9 |
| AK000258 | Hypothetical proteins | 0.27 | 0.07 | 14.0 | 11.1 | 7.9 | 16.1 | 6.2 | 18.9 |
| X89067 | Transient receptors | 0.20 | 0.14 | 3.7 | 2.2 | 2.4 | 2.6 | 8.0 | 18.1 |
| AL137619 | Is unknown | 0.16 | 0.08 | 6.3 | 6.7 | 10.8 | 10.5 | 7.9 | 16.5 |
| NM_003445 | Zinc finger proteins | 0.17 | 0.07 | 4.0 | 23.6 | 2.9 | 13.6 | 4.3 | 14.4 |
| X03084 | Complement component 1 | 0.36 | 0.15 | 2.4 | 3.1 | 2.9 | 7.7 | 3.4 | 13.7 |
| U27330 | Fucosyltransferase 5 | 0.39 | 0.08 | 2.4 | 2.5 | 2.6 | 12.1 | 3.5 | 13.0 |
| AF070549 | Is unknown | 0.16 | 0.09 | 2.7 | 4.7 | 7.9 | 10.3 | 4.2 | 12.6 |
| AB020335 | sel-1 sample | 0.19 | 0.24 | 2.9 | 2.6 | 2.0 | 7.3 | 4.7 | 12.4 |
| M26901 | Renin | 0.09 | 0.12 | 14.9 | 2.2 | 7.3 | 12.0 | 20.8 | 12.0 |
| Y07828 | Zinc finger proteins | 0.09 | 0.06 | 9.0 | 26.6 | 8.9 | 16.0 | 3.6 | 11.6 |
| AK001848 | Hypothetical proteins | 0.21 | 0.07 | 6.2 | 8.2 | 2.7 | 5.2 | 5.5 | 10.9 |
| NM_016331 | Zinc finger proteins | 0.16 | 0.08 | 7.6 | 5.1 | 7.0 | 25.5 | 5.5 | 10.9 |
| U75330 | Neural cell adhesion molecule 2 | 0.42 | 0.08 | 2.5 | 3.6 | 2.0 | 5.8 | 6.2 | 9.9 |
| AB037826 | Is unknown | 0.16 | 0.11 | 3.8 | 6.0 | 3.4 | 13.4 | 6.0 | 9.8 |
| M34041 | Adrenergic alpha-2B receptors | 0.30 | 0.13 | 4.5 | 4.5 | 3.7 | 8.6 | 5.6 | 9.8 |
| D38449 | Putative G protein-coupled receptors | 0.18 | 0.09 | 2.3 | 25.8 | 11.7 | 2.3 | 3.2 | 9.5 |
| AJ250562 | Transmembrane 4 superfamily member 2 | 0.13 | 0.10 | 10.0 | 8.4 | 2.2 | 8.1 | 16.3 | 9.1 |
| AK001807 | Hypothetical proteins | 0.18 | 0.12 | 4.2 | 5.3 | 4.6 | 3.2 | 4.0 | 8.3 |
| AL133051 | Is unknown | 0.09 | 0.07 | 5.1 | 13.6 | 6.0 | 9.1 | 2.2 | 8.2 |
| U43843 | Nerve-d 4 homolog | 0.61 | 0.10 | 2.0 | 6.4 | 2.3 | 16.6 | 2.2 | 8.1 |
| NM_013227 | Cartilage aggrecan 1 | 0.28 | 0.15 | 7.5 | 3.1 | 2.5 | 6.9 | 8.5 | 7.8 |
| AF226728 | Somatostatin receptor interacting proteins | 0.23 | 0.17 | 7.0 | 3.6 | 3.1 | 5.5 | 3.5 | 7.7 |
| AK001024 | Guanylic acid binding protein | 0.16 | 0.11 | 0.39 | 12.3 | 2.7 | 7.4 | 3.3 | 7.0 |
| AC002302 | Is unknown | 0.13 | 0.14 | 16.1 | 5.8 | 5.8 | 2.6 | 9.6 | 6.2 |
| AB007958 | Is unknown | 0.17 | 0.27 | 2.0 | 2.3 | 11.3 | 3.3 | 3.0 | 6.1 |
| AF059293 | CytokineReceptor-like factor 1 | 0.19 | 0.22 | 3.6 | 2.5 | 10.2 | 3.8 | 2.7 | 5.9 |
| V01512 | v-fos | 0.27 | 0.21 | 6.7 | 3.7 | 13.7 | 9.3 | 3.7 | 5.4 |
| U82762 | Sialyltransferase 8 | 0.23 | 0.15 | 3.2 | 6.5 | 2.7 | 9.2 | 5.7 | 5.4 |
| U44059 | Thyroid stimulating embryonic factor | 0.05 | 0.13 | 22.9 | 7.1 | 12.5 | 7.4 | 9.7 | 5.4 |
| X05323 | Antigens confirmed by monoclonal antibodies | 0.39 | 0.13 | 4.3 | 2.5 | 2.2 | 7.4 | 2.8 | 5.1 |
| U72671 | ICAM 5 | 0.25 | 0.14 | 5.3 | 2.7 | 3.7 | 10.0 | 3.2 | 4.8 |
| AL133626 | Hypothetical proteins | 0.26 | 0.25 | 2.2 | 4.2 | 2.9 | 3.0 | 2.6 | 4.7 |
| X96401 | MAX binding proteins | 0.31 | 0.29 | 6.9 | 2.3 | 4.9 | 3.1 | 2.9 | 4.6 |
| AL117533 | Is unknown | 0.05 | 0.26 | 8.2 | 2.7 | 11.1 | 2.5 | 11.9 | 4.5 |
| AK001550 | Hypothetical proteins | 0.10 | 0.30 | 8.0 | 2.0 | 4.9 | 2.1 | 7.8 | 4.5 |
| AB032436 | Human BNP1 mRNA | 0.14 | 0.21 | 5.1 | 2.2 | 9.1 | 4.5 | 6.4 | 4.4 |
| AL035447 | Hypothetical proteins | 0.28 | 0.23 | 4.3 | 3.7 | 8.7 | 5.2 | 3.7 | 4.2 |
| U09414 | Zinc finger proteins | 0.28 | 0.25 | 4.0 | 2.2 | 4.7 | 3.3 | 7.2 | 4.2 |
| AK001256 | Is unknown | 0.09 | 0.08 | 5.3 | 6.5 | 31.1 | 12.7 | 6.4 | 4.1 |
| L14813 | Carboxylate ligase like | 0.64 | 0.21 | 2.7 | 6.2 | 3.1 | 2.1 | 3.4 | 3.9 |
| AF038181 | Is unknown | 0.06 | 0.18 | 34.1 | 6.4 | 4.5 | 8.7 | 11.3 | 3.9 |
| NM_001486 | Glucokinase | 0.21 | 0.08 | 3.0 | 2.2 | 6.5 | 12.4 | 5.7 | 3.9 |
| AB033000 | Hypothetical proteins | 0.24 | 0.22 | 3.4 | 3.3 | 7.1 | 5.5 | 4.5 | 3.8 |
| AL117567 | DKFZP5660084 protein | 0.44 | 0.22 | 2.2 | 2.7 | 3.9 | 4.0 | 4.5 | 3.7 |
| NM_012126 | Saccharide sulfotransferase 5 | 0.31 | 0.20 | 5.5 | 5.4 | 3.8 | 5.5 | 2.6 | 3.5 |
| AL031687 | Is unknown | 0.16 | 0.27 | 5.9 | 2.6 | 3.4 | 2.3 | 4.9 | 3.5 |
| X04506 | Apolipoprotein B | 0.29 | 0.32 | 5.4 | 4.4 | 6.9 | 5.5 | 2.1 | 3.5 |
| NM_006641 | CCR9 | 0.35 | 0.11 | 3.3 | 3.3 | 2.2 | 16.5 | 2.3 | 3.5 |
| Y00970 | Acrosin | 0.12 | 0.14 | 8.2 | 8.8 | 3.1 | 6.2 | 17.5 | 3.4 |
| X67098 | rTS beta protein | 0.19 | 0.26 | 2.4 | 3.1 | 7.8 | 3.5 | 4.4 | 3.3 |
| U51990 | Pre-mRNA splicing factor | 0.56 | 0.19 | 2.2 | 3.0 | 2.8 | 13.7 | 2.9 | 3.0 |
| AF030555 | Fatty acid coenzyme A | 0.10 | 0.39 | 3.5 | 6.9 | 13.3 | 4.4 | 7.5 | 2.9 |
| AL009183 | TNFR superfamily, member 9 | 0.46 | 0.19 | 6.0 | 4.1 | 2.8 | 8.6 | 2.6 | 2.8 |
| AF045941 | sciellin | 0.16 | 0.21 | 11.6 | 2.4 | 2.8 | 2.2 | 4.1 | 2.8 |
| AF072756 | Kinase ankyrin 4 | 0.33 | 0.07 | 2.5 | 5.3 | 3.9 | 32.7 | 2.3 | 2.7 |
| X78678 | Hexanone kinase | 0.10 | 0.20 | 18.0 | 3.5 | 4.1 | 2.5 | 14.6 | 2.6 |
| AL031734 | Is unknown | 0.03 | 0.39 | 43.7 | 2.3 | 41.7 | 4.0 | 10.8 | 2.5 |
| D87717 | KIAA0013 Gene product | 0.35 | 0.42 | 4.2 | 2.3 | 3.6 | 2.6 | 2.9 | 2.5 |
| U01824 | Solute carrier family 1 | 0.42 | 0.29 | 4.8 | 2.3 | 4.2 | 7.1 | 4.2 | 2.4 |
| AF055899 | Solute carrier family 27 | 0.14 | 0.31 | 9.5 | 12.3 | 7.4 | 4.7 | 6.6 | 2.3 |
| U22526 | Lanosterol synthetase | 0.09 | 0.45 | 4.1 | 3.4 | 10.4 | 2.2 | 17.9 | 2.3 |
| AB032963 | Is unknown | 0.19 | 0.34 | 6.3 | 6.1 | 2.9 | 2.1 | 5.7 | 2.2 |
| NM_015974 | Lambda-crystallins | 0.17 | 0.25 | 11.4 | 2.8 | 5.9 | 2.4 | 5.8 | 2.2 |
| X82200 | Stimulated trans-acting factor | 0.23 | 0.15 | 8.2 | 3.4 | 3.0 | 2.8 | 11.3 | 2.2 |
| AL137522 | Is unknown | 0.12 | 0.26 | 12.1 | 3.7 | 12.6 | 6.9 | 4.3 | 2.2 |
| Z99916 | Crystallin, beta B3 | 0.28 | 0.65 | 2.5 | 2.1 | 3.6 | 2.2 | 2.6 | 2.1 |
| AF233442 | Ubiquitin-specific protease 21 | 0.41 | 0.31 | 2.6 | 3.6 | 3.6 | 4.5 | 3.4 | 2.1 |
| AK001927 | Hypothetical proteins | 0.24 | 0.52 | 7.6 | 5.6 | 5.0 | 2.5 | 4.1 | 2.0 |
[00121] Table 36: upregulation of polynucleotide expression in a549 cells induced by the peptide of formula F. A peptide concentration of 50. mu.g/ml was found to increase the expression of many polynucleotides. The peptides were incubated with human a549 epithelial cells for 4 hours, RNA was isolated, converted to labeled cDNA probes, and hybridized to a human operon array (PRHU 04). The intensity of the polynucleotide in the unstimulated control cells is shown in the third and fourth columns, which correspond to the cDNAs labeled with the dyes Cy3 and Cy5, respectively. "ratio ID #: control "column" refers to the intensity of polynucleotide expression in cells stimulated with peptide divided by the intensity of non-stimulated cells.
| Registration number | Gene | control-Cy 3 | control-Cy 5 | Ratio ID 40: control | Ratio ID 42: control | Ratio ID 43: control | Ratio ID 44: control | Ratio ID 45: control |
| AF025840 | Polymerase epsilon 2 | 0.34 | 0.96 | 3.4 | 2.0 | 2.0 | 2.1 | 4.3 |
| AF132495 | CGI-133 protein | 0.83 | 0.67 | 3.0 | 2.2 | 2.6 | 2.8 | 5.1 |
| AL137682 | Hypothetical proteins | 0.73 | 0.40 | 2.0 | 5.3 | 4.8 | 2.9 | 8.2 |
| U70426 | Modulators of G protein signaling 16 | 0.23 | 0.25 | 3.1 | 3.0 | 5.3 | 3.1 | 12.2 |
| AK001135 | Sec23 interacting protein p125 | 0.29 | 0.53 | 3.2 | 2.6 | 3.3 | 14.4 | 5.2 |
| AB023155 | KIAA0938 protein | 0.47 | 0.21 | 2.7 | 4.8 | 8.1 | 4.2 | 10.4 |
| AB033080 | Cell cycle process 8 protein | 0.31 | 0.31 | 4.4 | 2.2 | 5.9 | 4.3 | 6.9 |
| AF061836 | Ras-associated domain family 1 | 0.29 | 0.31 | 3.2 | 2.5 | 11.1 | 18.8 | 6.8 |
| AK000298 | Hypothetical proteins | 0.48 | 0.27 | 3.3 | 2.2 | 7.1 | 5.6 | 7.7 |
| L75847 | Zinc finger eggWhite colour (Bai) | 0.35 | 0.52 | 3.2 | 3.0 | 4.0 | 3.0 | 3.9 |
| X97267 | Protein tyrosine phosphatase | 0.19 | 0.24 | 4.1 | 9.3 | 2.4 | 4.2 | 8.3 |
| Z11933 | POU Domain 3 TF 2 | 0.09 | 0.23 | 8.7 | 2.5 | 3.6 | 4.3 | 8.2 |
| AB037744 | Is unknown | 0.37 | 0.57 | 2.6 | 2.9 | 2.7 | 3.0 | 3.1 |
| U90908 | Is unknown | 0.12 | 0.16 | 11.8 | 7.7 | 3.4 | 7.8 | 11.2 |
| AL050139 | Is unknown | 0.29 | 0.60 | 5.2 | 2.4 | 3.3 | 3.0 | 2.8 |
| AB014615 | Fibroblast growth factor 8 | 0.19 | 0.07 | 5.4 | 3.5 | 8.5 | 3.2 | 22.7 |
| M28825 | CD1A antigen | 0.51 | 0.36 | 4.1 | 2.6 | 2.0 | 4.6 | 4.4 |
| U27330 | Fucosyltransferase 5 | 0.39 | 0.08 | 3.3 | 2.1 | 24.5 | 8.2 | 19.3 |
| NM_006963 | Zinc finger proteins | 0.10 | 0.08 | 10.4 | 12.6 | 12.3 | 29.2 | 20.5 |
| AF093670 | Peroxisome biogenesis factor | 0.44 | 0.53 | 4.0 | 2.6 | 2.6 | 4.3 | 2.9 |
| AK000191 | Hypothetical proteins | 0.50 | 0.18 | 2.3 | 3.6 | 4.4 | 2.2 | 8.2 |
| AB022847 | Is unknown | 0.39 | 0.24 | 2.1 | 6.9 | 4.5 | 2.8 | 6.2 |
| AK000358 | Microfibrillar associated protein 3 | 0.28 | 0.28 | 5.7 | 2.0 | 3.5 | 5.2 | 5.2 |
| X74837 | Mannanase alpha class 1A | 0.10 | 0.07 | 13.1 | 18.4 | 23.6 | 16.3 | 20.8 |
| AF053712 | TNF superfamily member 11 | 0.17 | 0.08 | 11.3 | 9.3 | 13.4 | 10.6 | 16.6 |
| AL133114 | DKFZP586P2421 protein | 0.11 | 0.32 | 8.5 | 3.4 | 4.9 | 5.3 | 4.3 |
| AF049703 | E74-like factor 5 | 0.22 | 0.24 | 5.1 | 6.0 | 3.3 | 2.7 | 5.4 |
| AL137471 | Hypothetical proteins | 0.29 | 0.05 | 4.0 | 15.0 | 10.1 | 2.7 | 25.3 |
| AL035397 | Is unknown | 0.33 | 0.14 | 2.3 | 2.8 | 10.6 | 4.6 | 9.3 |
| AL035447 | Hypothetical proteins | 0.28 | 0.23 | 3.8 | 6.8 | 2.7 | 3.0 | 5.7 |
| X55740 | CD73 | 0.41 | 0.61 | 2.1 | 3.3 | 2.9 | 3.2 | 2.1 |
| NM_004909 | Paclitaxel resistance related gene 3 | 0.20 | 0.22 | 3.9 | 2.9 | 6.5 | 3.2 | 5.6 |
| AF233442 | Ubiquitin-specific protease | 0.41 | 0.31 | 2.9 | 4.7 | 2.7 | 3.5 | 3.9 |
| U92980 | Is unknown | 0.83 | 0.38 | 4.2 | 4.1 | 4.8 | 2.3 | 3.1 |
| AF105424 | Myosin heavy polypeptide-like | 0.30 | 0.22 | 2.8 | 3.3 | 4.4 | 2.3 | 5.3 |
| M26665 | histatin 3 | 0.29 | 0.26 | 7.9 | 3.5 | 4.6 | 3.5 | 4.5 |
| AF083898 | Ventral antigen 2 of neural tumor | 0.20 | 0.34 | 18.7 | 3.8 | 2.2 | 3.6 | 3.5 |
| AJ009771 | ariadne Drosophila homolog | 0.33 | 0.06 | 2.3 | 17.6 | 15.9 | 2.5 | 20.3 |
| AL022393 | Hypothetical protein P1 | 0.05 | 0.33 | 32.9 | 2.4 | 3.0 | 69.4 | 3.4 |
| AF039400 | Calcium-activated chloride channel family member 1 | 0.11 | 0.19 | 8.4 | 2.9 | 5.1 | 18.1 | 5.9 |
| AJ012008 | Dimethyl arginine dimethyl amino hydrolase | 0.42 | 0.43 | 5.1 | 3.3 | 3.2 | 6.2 | 2.6 |
| AK00542 | Hypothetical proteins | 0.61 | 0.24 | 2.1 | 4.5 | 5.0 | 3.7 | 4.4 |
| AL133654 | Is unknown | 0.27 | 0.40 | 2.8 | 2.1 | 2.5 | 2.5 | 2.6 |
| AL137513 | Is unknown | 0.43 | 0.43 | 6.4 | 3.2 | 3.8 | 2.3 | 2.3 |
| U05227 | GTP-binding proteins | 0.38 | 0.36 | 5.0 | 3.1 | 3.1 | 2.2 | 2.8 |
| D38449 | Putative G protein-coupled receptors | 0.18 | 0.09 | 5.8 | 6.7 | 6.7 | 9.1 | 10.4 |
| U80770 | Is unknown | 0.31 | 0.14 | 3.9 | 3.8 | 6.6 | 3.1 | 6.8 |
| X61177 | 1L-5 Rα | 0.40 | 0.27 | 2.6 | 4.4 | 9.8 | 8.1 | 3.6 |
| U35246 | Vesicle sorting protein 45A | 0.15 | 0.42 | 5.8 | 2.8 | 2.6 | 4.5 | 2.2 |
| AB017016 | Brain specific protein p25 alpha | 0.27 | 0.29 | 6.0 | 2.6 | 3.4 | 3.1 | 3.1 |
| X82153 | Cathepsin K | 0.45 | 0.20 | 4.2 | 5.2 | 4.8 | 4.4 | 4.6 |
| AC005162 | Most likely a carboxypeptidase precursor | 0.12 | 0.28 | 11.9 | 3.4 | 6.8 | 18.7 | 3.2 |
| AL137502 | Is unknown | 0.22 | 0.16 | 3.9 | 4.9 | 7.3 | 3.9 | 5.3 |
| U66669 | 3-hydroxyisobutyryl axial enzyme A hydrolase | 0.30 | 0.40 | 10.3 | 3.5 | 5.2 | 2.3 | 2.1 |
| AK000102 | Is unknown | 0.39 | 0.30 | 2.8 | 5.3 | 5.2 | 4.1 | 2.8 |
| AF034970 | Doxogenin 2 | 0.28 | 0.05 | 3.3 | 8.5 | 15.7 | 4.0 | 17.3 |
| AK000534 | Hypothetical proteins | 0.13 | 0.29 | 6.8 | 2.3 | 4.0 | 20.6 | 2.9 |
| J04599 | Biglycan proteoglycan | 0.39 | 0.30 | 4.0 | 3.7 | 4.0 | 4.8 | 2.8 |
| AL133612 | Is unknown | 0.62 | 0.33 | 2.7 | 3.4 | 5.2 | 3.0 | 2.5 |
| D10495 | Protein kinase C delta | 0.18 | 0.10 | 12.0 | 20.7 | 8.7 | 6.8 | 8.1 |
| X58467 | Cytochrome P450 | 0.07 | 0.24 | 15.4 | 4.7 | 7.9 | 34.4 | 3.4 |
| AF131806 | Is unknown | 0.31 | 0.25 | 2.6 | 3.4 | 5.7 | 7.0 | 3.2 |
| AK000351 | Hypothetical proteins | 0.34 | 0.13 | 4.0 | 6.9 | 5.5 | 2.8 | 6.3 |
| AF075050 | Hypothetical proteins | 0.55 | 0.09 | 2.7 | 17.8 | 5.1 | 2.2 | 8.3 |
| AK000566 | Hypothetical protein is unknown | 0.15 | 0.35 | 6.7 | 2.2 | 6.8 | 6.4 | 2.1 |
| U43328 | Cartilage connexin 1 | 0.44 | 0.19 | 2.5 | 6.2 | 6.9 | 7.8 | 3.8 |
| AF045941 | sciellin | 0.16 | 0.21 | 6.8 | 7.5 | 4.8 | 6.9 | 3.4 |
| U27655 | Modulators of G protein signaling 3 | 0.24 | 0.29 | 5.5 | 4.9 | 2.9 | 4.9 | 2.4 |
| AK000058 | Hypothetical proteins | 0.25 | 0.15 | 5.0 | 9.7 | 16.4 | 2.7 | 4.5 |
| AL035364 | Hypothetical proteins | 0.32 | 0.26 | 4.4 | 4.2 | 7.3 | 2.8 | 2.6 |
| AK001864 | Is unknown | 0.40 | 0.25 | 3.7 | 3.7 | 4.6 | 3.2 | 2.6 |
| AB015349 | Is unknown | 0.14 | 0.24 | 10.5 | 2.8 | 3.7 | 8.0 | 2.7 |
| V00522 | Class II MHC DR beta 3 | 0.62 | 0.22 | 4.8 | 3.9 | 4.7 | 2.5 | 3.0 |
| U75330 | Neural cell adhesion molecule 2 | 0.42 | 0.08 | 2.1 | 9.6 | 13.2 | 3.3 | 7.8 |
| NM_007199 | IL-1R-related kinase M | 0.15 | 0.25 | 8.7 | 7.8 | 8.6 | 16.1 | 2.5 |
| D30742 | Calcium/calmodulin-dependent protein kinase IV | 0.28 | 0.09 | 6.2 | 28.7 | 7.4 | 2.4 | 6.8 |
| X05978 | Cystatin A (cystatin A) | 0.63 | 0.17 | 2.7 | 4.8 | 9.4 | 2.2 | 3.6 |
| AF240467 | TLR-7 | 0.11 | 0.10 | 13.8 | 13.3 | 4.7 | 7.7 | 4.9 |
[00122] Table 37: upregulation of polynucleotide expression in a549 cells induced by peptides of formula G and other peptides. A peptide concentration of 50. mu.g/ml was found to increase the expression of many polynucleotides. The peptides were incubated with Human a549 epithelial cells for 4 hours, RNA was isolated, converted to labeled cDNA probes, and hybridized to a Human Operon array (PRHU 04). The intensity of the polynucleotide in the unstimulated control cells is shown in the second and third columns, which correspond to the cDNAs labeled with the dyes Cy3 and Cy5, respectively. "ratio ID #: control "column" refers to the intensity of polynucleotide expression in cells stimulated with peptide divided by the intensity of non-stimulated cells. Accession numbers and genes are expressed as: u00115, zinc finger protein; m91036, hemoglobin γ G; AK000070, hypothetical protein; AF055899, solute carrier family 27; AK001490, hypothetical protein; x97674, nuclear receptor coactivator 2; AB022847, unknown; AJ275986, a transcription factor; d10495, protein kinase C, δ; l36642, EphA 7; m31166, pentaxin-related gene; AF176012, unknown; AF072756, ankyrin 4; NM 014439, IL-1 superfamily z; AJ271351, a putative transcriptional regulator; AK000576, hypothetical protein; AJ272265, phosphoprotein 2 secretion; AL122038, hypothetical protein; AK000307, hypothetical protein; AB029001, KIAA1078 protein; u62437, cholinergic receptors; AF064854, unknown; AL031588, hypothetical protein; x89388, RAS p21 protein activator; d45399, phosphodiesterase; AB037716, hypothetical protein; x79981, cadherin 5; AF034208, RIG-like 7-1; AL133355, chromosome 21 open reading frame 53; NM — 016281, STE 20-like kinase; AF023614, transmembrane activator and CAML interacting protein; AF056717, as ash 2; AB029039, KIAA1116 protein; j03634, inhibin, β a; u80764, unknown; AB032963, unknown; x82835, type IX voltage-gated sodium ion channels.
| Registration number | control-Cy 3 | control-Cy 5 | ID 53: control | ID 54: control | ID 47: control | ID 48: control | ID 49: control | ID 50: control | ID 51: control | ID 52: control |
| U00115 | 0.51 | 0.07 | 27.4 | 7.3 | 2.4 | 3.1 | 4.8 | 8.3 | 3.5 | 20.0 |
| M91036 | 0.22 | 0.02 | 39.1 | 32.5 | 5.2 | 2.2 | 37.0 | 6.0 | 16.2 | 18.0 |
| AK000070 | 0.36 | 0.18 | 3.8 | 7.6 | 2.6 | 15.1 | 12.2 | 9.9 | 17.2 | 15.3 |
| AF055899 | 0.14 | 0.31 | 6.7 | 3.7 | 9.7 | 10.0 | 2.2 | 16.7 | 5.4 | 14.8 |
| AK001490 | 0.05 | 0.02 | 14.1 | 35.8 | 3.2 | 28.6 | 25.0 | 20.2 | 56.5 | 14.1 |
| X97674 | 0.28 | 0.28 | 3.2 | 3.7 | 4.0 | 10.7 | 3.3 | 3.1 | 4.0 | 13.2 |
| AB022847 | 0.39 | 0.24 | 4.1 | 4.4 | 4.5 | 2.7 | 3.7 | 10.4 | 5.0 | 11.3 |
| AJ275986 | 0.26 | 0.35 | 5.8 | 2.3 | 5.7 | 2.2 | 2.5 | 9.7 | 4.3 | 11.1 |
| D10495 | 0.18 | 0.10 | 8.0 | 3.4 | 4.6 | 2.0 | 6.9 | 2.5 | 12.7 | 10.3 |
| L36642 | 0.26 | 0.06 | 5.8 | 14.2 | 2.6 | 4.1 | 8.9 | 3.4 | 6.5 | 6.6 |
| M31166 | 0.31 | 0.12 | 4.8 | 3.8 | 12.0 | 3.6 | 9.8 | 2.4 | 8.8 | 6.4 |
| AF176012 | 0.45 | 0.26 | 3.1 | 2.9 | 2.8 | 2.6 | 2.3 | 6.9 | 3.0 | 5.8 |
| AF072756 | 0.33 | 0.07 | 9.9 | 9.3 | 4.4 | 4.3 | 3.2 | 4.9 | 11.9 | 5.4 |
| NM_014439 | 0.47 | 0.07 | 12.0 | 7.1 | 3.3 | 3.3 | 4.7 | 5.9 | 5.0 | 5.4 |
| AJ271351 | 0.46 | 0.12 | 3.4 | 3.5 | 2.3 | 4.7 | 2.3 | 2.7 | 6.9 | 5.2 |
| AK000576 | 0.27 | 0.06 | 7.4 | 15.7 | 2.9 | 4.7 | 9.0 | 2.4 | 8.2 | 5.1 |
| AJ272265 | 0.21 | 0.09 | 6.2 | 7.9 | 2.3 | 3.7 | 10.3 | 4.5 | 4.6 | 4.7 |
| AL122038 | 0.46 | 0.06 | 6.7 | 4.5 | 2.6 | 4.3 | 16.4 | 6.5 | 26.6 | 4.6 |
| AK000307 | 0.23 | 0.09 | 3.7 | 4.0 | 4.3 | 3.2 | 5.3 | 2.9 | 13.1 | 4.4 |
| AB029001 | 0.52 | 0.21 | 14.4 | 4.3 | 4.6 | 4.4 | 4.8 | 21.9 | 3.2 | 4.2 |
| U62437 | 0.38 | 0.13 | 12.6 | 6.5 | 4.2 | 6.7 | 2.2 | 3.7 | 4.8 | 3.9 |
| AF064854 | 0.15 | 0.16 | 2.6 | 2.9 | 6.2 | 8.9 | 14.4 | 5.0 | 9.1 | 3.9 |
| AL031588 | 0.40 | 0.26 | 8.3 | 5.2 | 2.8 | 3.3 | 5.3 | 9.0 | 5.6 | 3.4 |
| X89388 | 0.25 | 0.10 | 15.8 | 12.8 | 7.4 | 4.2 | 16.7 | 6.9 | 12.7 | 3.3 |
| D45399 | 0.21 | 0.18 | 3.0 | 4.7 | 3.3 | 4.4 | 8.7 | 5.3 | 5.1 | 3.3 |
| AB037716 | 0.36 | 0.40 | 5.1 | 7.5 | 2.6 | 2.1 | 3.5 | 3.1 | 2.4 | 2.8 |
| X79981 | 0.34 | 0.10 | 4.7 | 7.2 | 3.2 | 4.6 | 6.5 | 5.1 | 5.8 | 2.7 |
| AF034208 | 0.45 | 0.24 | 2.7 | 10.9 | 2.1 | 3.7 | 2.3 | 5.9 | 2.2 | 2.5 |
| AL133355 | 0.22 | 0.23 | 2.3 | 3.4 | 7.3 | 2.7 | 3.3 | 4.3 | 2.8 | 2.5 |
| NM_016281 | 0.40 | 0.19 | 6.6 | 10.6 | 2.1 | 2.8 | 5.0 | 11.2 | 10.6 | 2.5 |
| AF023614 | 0.11 | 0.42 | 2.2 | 2.2 | 6.0 | 7.5 | 5.0 | 2.7 | 2.0 | 2.4 |
| AF056717 | 0.43 | 0.62 | 4.3 | 3.2 | 5.1 | 4.0 | 4.6 | 9.7 | 3.1 | 2.2 |
| AB029039 | 0.79 | 0.49 | 2.7 | 3.3 | 3.7 | 2.0 | 2.3 | 2.4 | 4.8 | 2.2 |
| J03634 | 0.40 | 0.12 | 3.7 | 2.3 | 2.3 | 4.0 | 10.5 | 4.1 | 9.1 | 2.2 |
| U80764 | 0.31 | 0.18 | 2.3 | 7.4 | 4.2 | 2.3 | 5.1 | 3.3 | 8.8 | 2.1 |
| AB032963 | 0.19 | 0.34 | 4.0 | 7.3 | 5.0 | 3.0 | 2.9 | 6.7 | 3.8 | 2.1 |
| X82835 | 0.25 | 0.38 | 2.0 | 2.7 | 2.9 | 7.7 | 3.3 | 3.1 | 3.5 | 2.0 |
Example 5
Peptide-induced chemokines in cell lines, human whole blood and mice
[00123]The murine macrophage line RAW264.7, THP-1 cell (human monocyte) was usedHuman epithelial cell line (a549), human bronchial epithelial cells (16HBEo14) and human whole blood. HBE cells were grown in MEM (minimum essential medium) containing Erer's solution. THP-1 cells were grown and maintained in RPMI 1640 medium. RAW and a549 cell lines were maintained in DMEM supplemented with 10% fetal calf serum. These cells were seeded into 24-well plates with DMEM at a density of 10 per well6Individual cells (see above), a549 cells were seeded into 24-well plates with DMEM at a density of 10 per well5Cells (see above), all at 37 ℃ in 5% CO2Incubate overnight at medium temperature. DMEM was aspirated from overnight grown cells and replaced with fresh medium. After incubation of these cells with peptides, ELISA (R) was used&D Systems, Minneapolis, MN) to determine the chemokine release into the culture supernatant.
[00124] Animal studies were approved by the UBC animal management Committee (UBCACC # A01-0008). BALB/c mice were purchased from Charles River Laboratories and fed on standard animal facilities. Age, sex and weight matched adult mice were anesthetized by intraperitoneal injection of avermectin (4.4mM 2-2-2-tribromoethanol, 2.5% 2-methyl-2-butanol in distilled water) at a dose of 200 μ l per 10g body weight. Instillation was performed using a non-surgical intratracheal instillation method modified from Ho and Furst 1973. Briefly, the upper maxillary teeth of anesthetized rats were hooked on a wire at the top of a support frame to open their jaws and the chest was pushed with a spring to place their pharynx, larynx and trachea in a vertical line. The trachea is illuminated from the outside and an intubation tube is inserted into the clearly illuminated lumen of the trachea. Mu.l of peptide suspension or sterile water was placed in a small hole at the proximal end of the cannula and slowly instilled into the trachea with 200. mu.l of air. After instillation, the animals were held in an upright position for 2 minutes to allow the fluid to flow into the respiratory tree. After 4 hours, these mice were euthanized by intraperitoneal injection of 300mg/kg pentobarbital. The trachea is exposed, an intravenous catheter is inserted into the proximal end of the trachea, and tied appropriately with sutures. And (3) lavage is carried out: 0.75 ml of sterile PBS was introduced into the lungs through the tracheal cannula and after a few seconds the fluid was aspirated. This procedure was repeated three times with the same PBS sample. The lavage solution was placed in tubes and on ice, with a total recovery volume of approximately 0.5ml per mouse. This bronchoalveolar lavage (BAL) solution was centrifuged at 1200rpm for 10 minutes at high speed, the supernatant was removed, and TNF-. alpha.and MCP-1 were detected by ELISA.
[00125] The upregulation of chemokines by cationic peptides has been demonstrated in a number of different systems. Murine MCP-1 is a homolog of human MCP-1, which is a member of the β (C-C) chemokine family. MCP-1 has been shown to recruit monocytes, NK cells and some T lymphocytes. When human whole blood from RAW264.7 macrophages and 3 donors is enriched with increasing concentrations of the peptide SEQ ID NO: 1 stimulation, ELISA showed that they produced significant amounts of MCP-1 in their supernatant (Table 36). Stimulation of 24 h RAW264.7 cells with peptides at concentrations ranging from 20-50. mu.g/ml produced significant amounts of MCP-1 (200-400 pg/ml above background). When these cells (24 hours) and whole blood (4 hours) were stimulated with 100. mu.g/ml LL-37, high levels of MCP-1 were produced.
[00126] The effect of cationic peptides on the induction of chemokines was also examined in a completely different cell system, a549 human epithelial cells. Interestingly, although these cells produce MCP-1 in response to LPS, and this response is antagonized by the peptide; however, a549 cells respond directly to the peptide SEQID NO: at 1, no MCP-1 is produced. However, high concentrations of the peptide SEQ ID NO: 1 did induce the production of the neutrophil-specific chemokine IL-8 (table 37). Thus, SEQ ID NO: 1 are capable of inducing a range of responses at different concentrations and in different cell types. A number of peptides corresponding to each formula were tested for their ability to induce IL-8 in A549 cells (Table 38). Many of these peptides at low concentrations of 10. mu.g/ml induced IL-8 at background levels. High concentrations (100. mu.g/ml) of SEQ ID NO: 13 IL-8 was induced in whole blood (Table 39). Peptides of SEQ ID NO: 2 also induced IL-8 significantly.
[00127] BALB/c mice were given by intratracheal instillation SEQ ID NO: MCP-1 and TNF-alpha levels in bronchoalveolar lavage fluid were measured after 3-4 hours in water with or without endotoxin. We found that the peptide SEQ ID NO: 1 the former produced significantly higher levels of MCP-1 than mice given either water or anesthetic alone (Table 42). For the peptide SEQ ID NO: 1 did not have a pro-inflammatory response because the peptide did not induce significantly more TNF-alpha than mice given water or anesthetic alone. It was also found that the peptide SEQ ID NO: 1 (up to 100 μ g/ml) treated RAW264.7 cells and bone marrow derived macrophages, peptide SEQ ID NO: 1 did not significantly induce TNF-. alpha.production (Table 43). Thus, the peptide SEQ ID NO: 1 selectively induces the production of chemokines without inducing the production of inflammatory mediators such as TNF-alpha. This demonstrates that the peptide SEQ ID NO: 1 has a dual role, both as a factor capable of preventing bacterial product-induced inflammation and helping to recruit phagocytic cells that can clear infection.
[00128] Table 38: induction of MCP-1 in RAW264.7 cells and human whole blood. RAW264.7 murine macrophages and human whole blood were stimulated with increasing concentrations of LL-37 for 4 hours. Human whole blood samples were centrifuged at high speed, the serum was removed, and MCP-1 was detected in ELISA, while MCP-1 was detected in the supernatant of RAW264.7 cells in ELISA. Data for RAW cells are expressed as mean ± standard deviation of three or more experiments, and data for human whole blood are expressed as mean ± standard deviation from three different donors.
| Peptide, SEQ ID NO: 1(μ g/ml) | Monocyte chemistry inducing protein (MCP) -1(pg/ml)* | |
| RAW cells | Whole blood | |
| 0 | 135.3±16.3 | 112.7±43.3 |
| 10 | 165.7±18.2 | 239.3±113.3 |
| 50 | 367±11.5 | 371±105 |
| 100 | 571±17.4 | 596±248.1 |
[00129] Table 39: induction of IL-8 in a549 cells and human whole blood. A549 cells and human whole blood were stimulated with increasing concentrations of peptide for 24 hours and 4 hours, respectively. Human whole blood samples were centrifuged at high speed, the serum was removed, and IL-8 was detected in ELISA, while IL-8 was detected in the supernatant of A549 cells in ELISA. Data for a549 cells are expressed as mean ± standard deviation of three or more experiments, and data for human whole blood are expressed as mean ± standard deviation from three different donors.
| Peptide, SEQ ID NO: 1(μ g/ml) | IL-8(pg/ml) | |
| A549 cell | Whole blood | |
| 0 | 172±29.1 | 660.7±126.6 |
| 1 | 206.7±46.1 | |
| 10 | 283.3±28.4 | 945.3±279.9 |
| 20 | 392±31.7 | |
| 50 | 542.3±66.2 | 1160.3±192.4 |
| 100 | 1175.3±188.3 | |
[00130] Table 40: the cationic peptide in A549 cells induced IL-8. A549 human epithelial cells were stimulated with 10 μ g of peptide for 24 hours. The supernatant was removed and IL-8 was detected by ELISA.
| Peptide (10. mu.g/ml) | IL-8(ng/ml) |
| Without peptides | 0.164 |
| LPS, peptide free | 0.26 |
| SEQ ID NO:1 | 0 278 |
| SEQ ID NO:6 | 0.181 |
| SEQ ID NO:7 | 0.161 |
| SEQ ID NO:9 | 0.21 |
| SEQ ID NO:10 | 0.297 |
| SEQ ID NO:13 | 0.293 |
| SEQ ID NO:14 | 0.148 |
| SEQ ID NO:16 | 0.236 |
| SEQ ID NO:17 | 0.15 |
| SEQ ID NO:19 | 0.161 |
| SEQ ID NO:20 | 0.151 |
| SEQ ID NO:21 | 0.275 |
| SEQ ID NO:22 | 0.314 |
| SEQ ID NO:23 | 0.284 |
| SEQ ID NO:24 | 0.139 |
| SEQ ID NO:26 | 0.201 |
| SEQ ID NO:27 | 0.346 |
| SEQ ID NO:28 | 0.192 |
| SEQ ID NO:29 | 0.188 |
| SEQ ID NO:30 | 0.284 |
| SEQ ID NO:31 | 0.168 |
| SEQ ID NO:33 | 0.328 |
| SEQ ID NO:34 | 0.315 |
| SEQ ID NO:35 | 0.301 |
| SEQ ID NO:36 | 0.166 |
| SEQ ID NO:37 | 0.269 |
| SEQ ID NO:38 | 0.171 |
| SEQ ID NO:40 | 0.478 |
| SEQ ID NO:41 | 0.371 |
| SEQ ID NO:42 | 0.422 |
| SEQ ID NO:43 | 0.552 |
| SEQ ID NO:44 | 0.265 |
| SEQ ID NO:45 | 0.266 |
| SEQ ID NO:47 | 0.383 |
| SEQ ID NO:48 | 0.262 |
| SEQ ID NO:49 | 0.301 |
| SEQ ID NO:50 | 0.141 |
| SEQ ID NO:51 | 0.255 |
| SEQ ID NO:52 | 0.207 |
| SEQ ID NO:53 | 0.377 |
| SEQ ID NO:54 | 0.133 |
[00131] Table 41: peptides in human blood induce IL-8. Human whole blood was stimulated with increasing concentrations of peptide for 4 hours. Human blood samples were centrifuged, the serum removed, and IL-8 detected by ELISA.
Data are from the mean of two donors.
| SEQ ID NO:3(μg/ml) | IL-8(pg/ml) |
| 0 | 85 |
| 10 | 70 |
| 100 | 323 |
[00132] Table 42: induction of IL-8 in HBE cells. Increasing concentrations of peptide were incubated with HBE cells for 8 hours, the supernatant removed, and IL-8 detected by ELISA. Data are presented as mean ± standard deviation of three or more experiments.
| SEQ ID NO:2(μg/ml) | IL-8(pg/ml) |
| 0 | 552±90 |
| 0.1 | 670±155 |
| 1 | 712±205 |
| 10 | 941±15 |
| 50 | 1490±715 |
[00133] Table 43: induction of IL-8 in undifferentiated THP-1 cells. The peptides at the indicated concentrations were incubated with human monocyte THP-1 cells for 8 hours, the supernatant removed, and IL-8 detected by ELISA.
| SEQ ID NO:3(μg/ml) | IL-8(pg/ml) |
| 0 | 10.6 |
| 10 | 17.2 |
| 50 | 123.7 |
[00134] Table 44: in the mouse airway the peptide SEQ ID NO: 1 induces MCP-1. BALB/c mice were anesthetized with Avertine and either instilled with peptide or water intratracheally or without instillation (no treatment). Mice were monitored for 4 hours and BAL fluid was isolated and assayed for MCP-1 and TNF-. alpha.concentrations by ELISA. Data are presented as mean ± standard deviation of four or five mice for each condition.
| Condition | MCP-1(pg/ml) | TNF-α(pg/ml) |
| Water (W) | 16.5±5 | 664±107 |
| Peptides | 111±30 | 734±210 |
| Avertin (avertin) | 6.5±0.5 | 393±129 |
[00135] Table 45: the cationic peptide did not significantly induce TNF- α. The indicated peptide (40. mu.g/ml) was incubated with RAW 246.7 macrophages for 6 hours. Supernatants were collected and assayed for TNF-. alpha.levels by ELISA. Data are presented as mean ± standard deviation of three or more experiments.
| Peptide treatment | TNF-α(pg/ml) |
| Background of the culture Medium | 56±8 |
| LPS treatment, no peptide | 15207±186 |
| SEQ ID NO:1 | 274±15 |
| SEQ ID NO:5 | 223±45 |
| SEQ ID NO:6 | 297±32 |
| SEQ ID NO:7 | 270±42 |
| SEQ ID NO:8 | 166±23 |
| SEQ ID NO:9 | 171±33 |
| SEQ ID NO:10 | 288±30 |
| SEQ ID NO:12 | 299±65 |
| SEQ ID NO:13 | 216±42 |
| SEQ ID NO:14 | 226±41 |
| SEQ ID NO:15 | 346±41 |
| SEQ ID NO:16 | 341±68 |
| SEQ ID NO:17 | 249±49 |
| SEQ ID NO:19 | 397±86 |
| SEQ ID NO:20 | 285±56 |
| SEQ ID NO:21 | 263±8 |
| SEQ ID NO:22 | 195±42 |
| SEQ ID NO:23 | 254±58 |
| SEQ ID NO:24 | 231±32 |
| SEQ ID NO:26 | 281±34 |
| SEQ ID NO:27 | 203±42 |
| SEQ ID NO:28 | 192±26 |
| SEQ ID NO:29 | 242±40 |
| SEQ ID NO:31 | 307±71 |
| SEQ ID NO:33 | 196±42 |
| SEQ ID NO:34 | 204±51 |
| SEQ ID NO:35 | 274±76 |
| SEQ ID NO:37 | 323±41 |
| SEQ ID NO:38 | 199±38 |
| SEQ ID NO:43 | 947±197 |
| SEQ ID NO:44 | 441±145 |
| SEQ ID NO:45 | 398±90 |
| SEQ ID NO:48 | 253±33 |
| SEQ ID NO:49 | 324±38 |
| SEQ ID NO:50 | 311±144 |
| SEQ ID NO:53 | 263±40 |
| SEQ ID NO:54 | 346±86 |
Example 6
Cationic peptides increase surface expression of chemokine receptors
[00136] To analyze cell surface expression of IL-8RB, CXCR-4, CCR2 and LFA-1, RAW macrophages were stained with 10 μ g/ml of the appropriate primary antibody (Santa Cruz Biotechnology) followed by FITC-conjugated goat anti-rabbit IgG [ IL-8RB and CXCR-4(Jackson ImmunoResearch Laboratories, West Grove, Pa) ] or FITC-conjugated Sandonkey anti-goat IgG (Santa Cruz) ]. Cells were analyzed by FACscan, counting 10,000 times and turning on the front and side diffusers to exclude cell debris.
[00137] Polynucleotide array data indicate that some peptides upregulate expression of the chemokine receptors IL-8RB, CXCR-4, and CCR2 by 10, 4, and 1.4 fold, respectively, compared to unstimulated cells. To confirm the polynucleotide array data, surface expression of the receptor on RAW cells stimulated with the peptide for 4 hours was detected by flow cytometry. When 50 μ g/ml of peptide was incubated with RAW cells for 4 hours, IL-8RB was upregulated on average 2.4-fold over unstimulated cells, CXCR-4 was upregulated on average 1.6-fold over unstimulated cells, and CCR2 was upregulated 1.8-fold over unstimulated cells (Table 46). As a control, CEMA was shown to result in similar up-regulation. Bac2A was the only peptide that could significantly up-regulate LFA-1 (3.8 fold higher than control cells).
[00138] Table 46: in response to the peptide, the surface expression of CXCR-4, IL-8RB and CCR2 was increased. RAW macrophages were stimulated with the peptide for 4 hours. The cells were washed and stained with appropriate primary and FITC-labeled secondary antibodies. Data shown represent mean (fold change in RAW cells stimulated with peptide) ± standard deviation.
| Peptides | Concentration (μ g/ml) | Fold increase in protein expression | ||
| IL-8RB | CXCR-4 | CCR2 | ||
| SEQ IDNO:1 | 10 | 1.0 | 1.0 | 1.0 |
| SEQ IDNO:1 | 50 | 1.3±0.05 | 1.3±0.03 | 1.3±0.03 |
| SEQ IDNO:1 | 100 | 2.4±0.6 | 1.6±0.23 | 1.8±0.15 |
| SEQ IDNO:3 | 100 | 2.0±0.6 | Do not make | 4.5 |
| CEMA | 50 | 1.6±0.1 | 1.5±02 | 1.5±0.15 |
| 100 | 3.6±0.8 | Do not make | 4.7±1.1 |
Example 7
Cationic peptide induced phosphorylation of MAP kinase
[00139]At 2.5X 105-5×105Cells were seeded at a density of one cell/ml and allowed to stand overnight. In the morning, cells were washed once with serum-free medium (serum-free medium-4 hours). The medium was removed and replaced with PBS, and then left at 37 ℃ for 15 minutes and at room temperature for 15 minutes. Peptides (at a concentration of 0.1. mu.g/ml to 50. mu.g/ml) or water were added and incubated for 10 minutes. PBS was removed quickly and replaced with ice cold Radioimmunoprecipitation (RIPA) buffer and inhibitors (NaF, B-glycerophosphate, MOL, vanadate, PMSF, leupeptin, aprotininLeupeptin). The plates were shaken on ice for 10-15 minutes or until the cells were lysed and the lysate was collected. For THP-1 cells, the procedure was slightly different; more cells (5X 10) were used6). They were left overnight in the absence of serum, 1ml of ice-cold PBS was added to stop the reaction, then placed on ice for 5-10 minutes, spun down, and then resuspended with RIPA. Protein concentration was determined using protein assay (Pierce, Rockford, IL.). Cell lysates (20. mu.g protein) were separated by SDS-PAGE and transferred to nitrocellulose membranes. The membrane was blocked with 10mM Tris-HCl, pH7.5, 150mM NaCl (TBS)/5% skimmed milk powder for 1 hour, then incubated overnight in cold TBS/0.05% Tween 20 containing primary antibody. After washing with TBS/0.05% Tween 20 for 30 minutes, it was incubated for another 1 hour at room temperature with goat anti-mouse IgG conjugated with horseradish peroxidase (1: 10,000 in TBS/0.05% Tween 20). Immunoreactive bands were visualized using Enhanced Chemiluminescence (ECL) detection after washing the membranes with TBS/0.1% Tween 20 for 30 minutes. For experiments using peripheral blood mononuclear cells: peripheral blood (50-100ml) was collected from all individuals. Monocytes were isolated from peripheral blood by density gradient centrifugation on Ficoll-Hypaque. Interphase cells (monocytes) were recovered, washed, and then resuspended in the recommended primary medium for cell culture (RPMI-1640) containing 10% Fetal Calf Serum (FCS) and 1% L-glutamine. At a rate of 4X 10 per hole6Density of Individual cells were loaded into 6-well plates at 37 ℃ in 5% CO2The mixture was left in the air for 1 hour to allow adhesion to occur. The surface-floating medium and non-adherent cells were washed away and the appropriate medium and peptide were added. It was estimated from their ability to exclude trypan blue that > 99% of freshly harvested cells should survive. After stimulation with the peptide, the cells were lysed with RIPA buffer in the presence of various phosphatase inhibitors and kinase inhibitors, and the lysates were collected. Protein content was analyzed and approximately 30. mu.g of each sample was loaded onto a 12% SDS-PAGE gel. Proteins in the gel were transferred to nitrocellulose and blocked with Tris Buffered Saline (TBS) containing 5% skim milk powder and 1% Triton X100 for 1 hour. Phosphorylation was detected with phosphorylation-specific antibodies.
[00140] The results of peptide-induced phosphorylation are summarized in table 46. It was found that in murine macrophage RAW cell line and HBE cells, SEQ ID NO: 2 caused dose-dependent phosphorylation of p38 and ERK 1/2. In the THP-1 human monocyte cell line, SEQ ID NO: 3 leads to phosphorylation of MAP kinase, in murine RAW cell line, SEQ ID NO: 3 leads to phosphorylation of ERK 1/2.
[00141] Table 47: phosphorylation of MAP kinase in response to peptides.
| Cell lines | Peptides | Phosphorylated by MAP kinase | |
| p38 | ERK1/2 | ||
| RAW 264.7 | SEQ ID NO:3 | - | + |
| SEQ ID NO:2 | + | + | |
| HBE | SEQ ID NO:3 | + | |
| SEQ ID NO:2 | + | + | |
| THP-1 | SEQ ID NO:3 | + | + |
| SEQ ID NO:2 | |||
[00142] Table 48: peptide phosphorylation of MAP kinase (SEQ ID NO: 1, 50. mu.g/ml) in human blood mononuclear cells was used to promote phosphorylation.
| P38 phosphorylation | ERK1/2 phosphorylation | ||
| 15 minutes | 60 minutes | 15 minutes | 60 minutes |
| + | - | + | + |
Example 8
Cationic peptides defend against bacterial infections by boosting immune responses
[00143]BALB/c mice were given 1X 10 injections intraperitoneally5Salmonella and cationic peptide (200 μ g). Mice were monitored for 24 hours when they died, spleens were removed, homogenized, resuspended in PBS, and plated on Luria Broth agar plates containing kanamycin (50. mu.g/ml). The plates were incubated overnight at 37 ℃ and the surviving bacteria were counted (tables 49 and 50). CD-1 mice were administered by intraperitoneal injection containing 1X 1085% porcine mucin fluid of Staphylococcus aureus and cationic peptide (200. mu.g) (Table 51). The mice were monitored for 3 days, at which time they died, blood was removed, plated, and the number of viable cells was counted. CD-1 male mice were administered 5.8X 10 by Intraperitoneal (IP) injection6CFU EHEC bacteria and cationic peptide (200 μ g) were monitored for 3 days (table 52). In each of these animal models a fraction of the peptides showed protection against infection. When the results of the defense analysis in tables 49 and 50 were compared to the results of gene expression in tables 31-37, it was found that the most defensive peptides in the salmonella model were able to induce a common set of genes in epithelial cells (table 53). This clearly indicates that the pattern of gene expression is consistent with the ability of the peptide to display defense. The results of the Minimum Inhibitory Concentration (MIC) test (table 54) indicate that many of these cationic peptides are not directly resistant to microorganisms. This suggests that the ability of a peptide to defend against infection is dependent on the ability of the peptide to stimulate the host's innate immunity, rather than on direct antimicrobial activity.
[00144] Table 49: effect of cationic peptide on Salmonella infection in BALB/c mice. Salmonella and peptide were injected intraperitoneally into BALB/c mice, and after 24 hours, these animals died painlessly, spleens were removed, homogenized, diluted with PBS, plates counted, and bacterial survival was determined.
| Peptide treatment | Viable bacteria in the spleen (CFU/ml) | Statistical significance (p value) |
| Control | 2.70±0.84X105 | |
| SEQ ID NO:1 | 1.50±0.26X105 | 0.12 |
| SEQ ID NO:6 | 2.57±0.72X104 | 0.03 |
| SEQ ID NO:13 | 3.80±0.97X104 | 0.04 |
| SEQ ID NO:17 | 4.79±1.27X104 | 0.04 |
| SEQ ID NO:27 | 1.01±0.26X105 | 0.06 |
[00145] Table 50: effect of cationic peptide on Salmonella infection in BALB/c mice. Salmonella and peptide were injected intraperitoneally into BALB/c mice, and after 24 hours, these animals died painlessly, spleens were removed, homogenized, diluted with PBS, plates counted, and bacterial survival was determined.
| Peptide treatment | Viable bacteria in the spleen (CFU/ml) |
| Control | 1.88±0.16X104 |
| SEQ ID NO:48 | 1.98±0.18X104 |
| SEQ ID NO:26 | 7.1±1.37X104 |
| SEQ ID NO:30 | 5.79±0.43X103 |
| SEQ ID NO:37 | 1.57±0.44X104 |
| SEQ ID NO:5 | 2.75±0.59X104 |
| SEQ ID NO:7 | 5.4±0.28X103 |
| SEQ ID NO:9 | 1.23±0.87X104 |
| SEQ ID NO:14 | 2.11±0.23X103 |
| SEQ ID NO:20 | 2.78±0.22X104 |
| SEQ ID NO:23 | 6.16±0.32X104 |
[00146]Table 51: effect of cationic peptide in murine model of Staphylococcus aureus infection. Will contain 1X 108Bacterial 5% porcine mucin solution was injected Intraperitoneally (IP) into CD-1 mice. Cationic peptide (200 μ g) was administered via a single intraperitoneal injection. After 3 days of monitoring, the rats died painlessly, blood was removed and plated to count the number of survivors. The following peptides were not effective in controlling staphylococcus aureus infections: SEQ ID NO: 48, SEQ ID NO: 26.
| treatment of | CFU/ml (blood) | # surviving mice (3 days)/Total number of mice in this group |
| Without peptides | 7.61±1.7X103 | 6/8 |
| SEQ ID NO:1 | 0 | 4/4 |
| SEQ ID NO:27 | 2.25±0.1X102 | 3/4 |
| SEQ ID NO:30 | 1.29±0.04X102 | 4/4 |
| SEQ ID NO:37 | 9.65±0.41X102 | 4/4 |
| SEQ ID NO:5 | 3.28±1.7X103 | 4/4 |
| SEQ ID NO:6 | 1.98±0.05X102 | 3/4 |
| SEQ ID NO:7 | 3.8±0.24X103 | 4/4 |
| SEQ ID NO:9 | 2.97±0.25X102 | 4/4 |
| SEQ ID NO:13 | 4.83±0.92X103 | 3/4 |
| SEQ ID NO:17 | 9.6±0.41X102 | 4/4 |
| SEQ ID NO:20 | 3.41±1.6X103 | 4/4 |
| SEQ ID NO:23 | 4.39±2.0X103 | 4/4 |
[00147]Table 52: effect of cationic peptides in EHEC infection model in mice. Mixing 5.8X 106CFU EHEC bacteria were injected Intraperitoneally (IP) into CD-1 males (5 weeks old). Cationic peptide (200 μ g) was administered via a single intraperitoneal injection. These mice were monitored for 3 days.
| Treatment of | Peptides | Survival (%) |
| Control | Is free of | 25 |
| SEQ ID NO:23 | 200μg | 100 |
[00148] Table 53: peptides active in vivo induce upregulation of gene expression patterns in a549 epithelial cells. It was found that the peptide of SEQ ID NO: 30. SEQ ID NO: 7 and SEQ ID NO: 13 after 4 hours of treatment, each peptide increased the expression of a set of genes. The peptides were incubated with Human a549 epithelial cells for 4 hours, RNA was isolated, converted to labeled cDNA probes, and hybridized to a Human Operon array (PRHU 04). The intensity of the polynucleotide in unstimulated control cells is shown in the second column (average of both cases of labeling cDNA with Cy3 and Cy 5). Fold up of upregulation one column refers to the intensity of polynucleotide expression in cells stimulated with peptide divided by the intensity of non-stimulated cells. The table also includes peptides SEQ ID NO: 37, which is not active in a murine infection model.
| Target (registration number) | Unstimulated cell strength | Fold up regulation of gene expression relative to untreated cells |
| SEQ IDNO:30 | SEQ IDNO:7 | SEQ IDNO:13 | SEQ IDNO:37 | ||
| Zinc finger protein (AF061261) | 13 | 2.6 | 9.4 | 9.4 | 1.0 |
| Cell cycle gene (S70622) | 1.62 | 8.5 | 3.2 | 3.2 | 0.7 |
| IL-10 receptor (U00672) | 0.2 | 2.6 | 9 | 4.3 | 0.5 |
| Transferase (AF038664) | 0.09 | 12.3 | 9.7 | 9.7 | 0.1 |
| Homeobox protein (AC004774) | 0.38 | 3.2 | 2.5 | 2.5 | 1.7 |
| Bifurcate head type protein (AF042832) | 0.17 | 14.1 | 3.5 | 3.5 | 0.9 |
| Unknown (AL096803) | 0.12 | 4.8 | 4.3 | 4.3 | 0.6 |
| KIAA0284 protein (AB006622) | 0.47 | 3.4 | 2.1 | 2.1 | 1.3 |
| Hypothetical protein (AL022393) | 0.12 | 4.4 | 4.0 | 4.0 | 0.4 |
| Receptor (AF112461) | 0.16 | 2.4 | 10.0 | 10.0 | 1.9 |
| Hypothetical protein (AK002104) | 0.51 | 4.7 | 2.6 | 2.6 | 1.0 |
| Protein (AL050261) | 0.26 | 3.3 | 2.8 | 2.8 | 1.0 |
| Polypeptide (AF105424) | 0.26 | 2.5 | 5.3 | 5.3 | 1.0 |
| SPR1 protein (AB031480) | 0.73 | 3.0 | 2.7 | 2.7 | 1.3 |
| Dehydrogenase (D17793) | 4.38 | 2.3 | 2.2 | 2.2 | 0.9 |
| Transferase (M63509) | 0.55 | 2.7 | 2.1 | 2.1 | 1.0 |
| Peroxisome factor (AB013818) | 0.37 | 3.4 | 2.9 | 2.9 | 1.4 |
[00149] Table 54: most cationic peptides studied here, and in particular those effective in infection models, are not significantly antimicrobial. Serial dilutions of peptides were incubated with the indicated bacteria in 96-well plates overnight. The lowest concentration of peptide that killed the bacteria was expressed as the MIC. Symbol > indicates that the MIC is too large to measure. MICs of 8. mu.g/ml or less are considered clinically significant activities. Abbreviations: coli, Escherichia coli (Escherichia coli); s. aureus, Staphylococcus aureus (Staphylococcus aureus); p. aerug, Pseudomonas aeruginosa (Pseudomonas aeruginosa); typhim, salmonella typhimurium (salmonella spp.), c. rhod (citrobacter rhodens); EHEC, enterohemorrhagic escherichia coli (Enterohaemorrhagic e.
| Peptides | MIC(μg/ml) | |||||
| E.coli | S.aureus | P.aerug | S.typhim | C.rhod | EHEC | |
| Polymyxin | 0.25 | 16 | 0.25 | 0.5 | 0.25 | 0.5 |
| Gentamicin | 0.25 | 0.25 | 0.25 | 0.25 | 0.25 | 0.5 |
| SEQ ID NO:1 | 32 | > | 96 | 64 | 8 | 4 |
| SEQ ID NO:5 | 128 | > | > | > | 64 | 64 |
| SEQ ID NO:6 | 128 | > | > | 128 | 64 | 64 |
| SEQ ID NO:7 | > | > | > | > | > | > |
| SEQ ID NO:8 | > | > | > | > | > | > |
| SEQ ID NO:9 | > | > | > | > | > | > |
| SEQ ID NO:10 | > | > | > | > | > | 64 |
| SEQ ID NO:12 | > | > | > | > | > | > |
| SEQ ID NO:13 | > | > | > | > | > | > |
| SEQ ID NO:14 | > | > | > | > | > | > |
| SEQ ID NO:15 | 128 | > | > | > | 128 | 64 |
| SEQ ID NO:16 | > | > | > | > | > | > |
| SEQ ID NO:17 | > | > | > | > | > | > |
| SEQ ID NO:19 | 8 | 16 | 16 | 64 | 4 | 4 |
| SEQ ID NO:2 | 4 | 16 | 32 | 16 | 64 | |
| SEQ ID NO:20 | 8 | 8 | 8 | 8 | 16 | 8 |
| SEQ ID NO:21 | 64 | 64 | 96 | 64 | 32 | 32 |
| SEQ ID NO:22 | 8 | 12 | 24 | 8 | 4 | 4 |
| SEQ ID NO:23 | 4 | 8 | 8 | 16 | 4 | 4 |
| SEQ ID NO:24 | 16 | 16 | 4 | 16 | 16 | 4 |
| SEQ ID NO:26 | 0.5 | 32 | 64 | 2 | 2 | 0.5 |
| SEQ ID NO:27 | 8 | 64 | 64 | 16 | 2 | 4 |
| SEQ ID NO:28 | > | > | > | 64 | 64 | 128 |
| SEQ ID NO:29 | 2 | > | > | 16 | 32 | 4 |
| SEQ ID NO:30 | 16 | > | 128 | 16 | 16 | 4 |
| SEQ ID NO:31 | > | > | 128 | > | > | 64 |
| SEQ ID NO:33 | 16 | 32 | > | 16 | 64 | 8 |
| SEQ ID NO:34 | 8 | > | > | 32 | 64 | 8 |
| SEQ ID NO:35 | 4 | 128 | 64 | 8 | 8 | 4 |
| SEQ ID NO:36 | 32 | > | > | 32 | 32 | 16 |
| SEQ ID NO:37 | > | > | > | > | > | > |
| SEQ ID NO:38 | 0.5 | 32 | 64 | 4 | 8 | 4 |
| SEQ ID NO:40 | 4 | 32 | 8 | 4 | 4 | 2 |
| SEQ ID NO:41 | 4 | 64 | 8 | 8 | 2 | 2 |
| SEQ ID NO:42 | 1.5 | 64 | 4 | 2 | 2 | 1 |
| SEQ ID NO:43 | 8 | 128 | 16 | 16 | 8 | 4 |
| SEQ ID NO:44 | 8 | > | 128 | 128 | 64 | 64 |
| SEQ ID NO:45 | 8 | > | 128 | 128 | 16 | 16 |
| SEQ ID NO:47 | 4 | > | 16 | 16 | 4 | 4 |
| SEQ ID NO:48 | 16 | > | 128 | 16 | 1 | 2 |
| SEQ ID NO:49 | 4 | > | 16 | 8 | 4 | 4 |
| SEQ ID NO:50 | 8 | > | 16 | 16 | 16 | 8 |
| SEQ ID NO:51 | 4 | > | 8 | 32 | 4 | 8 |
| SEQ ID NO:52 | 8 | > | 32 | 8 | 2 | 2 |
| SEQ ID NO:53 | 4 | > | 8 | 8 | 16 | 8 |
| SEQ ID NO:54 | 64 | > | 16 | 64 | 16 | 32 |
Example 9
Use of polynucleotides induced by bacterial signaling molecules in diagnosis/screening
[00150] Salmonella typhimurium LPS and E coli 0111: b4LPS was purchased from sigma chemical Co (st. LTA (Sigma) from Staphylococcus aureus was resuspended in endotoxin-free water (Sigma). The Limulus amoebocyte lysate test (Sigma) was performed on LTA preparations to confirm that they were not significantly contaminated with endotoxin (i.e., less than 1ng/nl, a concentration that did not result in significant cytokine production by RAW cells). CpG oligodeoxynucleotides were synthesized using a Model 392DNA/RNA synthesizer from Applied biosystems (Applied biosystems, Missisaga, ON.), purified and resuspended in endotoxin-free water (Sigma). The following sequences CpG were used: 5'-TCATGACGTTCCTGACGTT-3' (SEQ ID NO: 57) and non-CpG: 5'-TTCAGGACTTTCCTCAGGTT-3' (SEQ ID NO: 58). The ability of the non-CpG oligomer to stimulate cytokine production was tested and found to not result in significant production of TNF- α or IL-6, and thus can be considered a negative control. RAW264.7 cells were incubated with medium alone, 100ng/ml Salmonella typhimurium LPS, 1. mu.g/ml Staphylococcus aureus LTA or 1. mu.M CpG for 4 hours (these concentrations optimally induced tumor necrosis factor (TNF-. alpha.) production by RAW cells), and RNA was isolated from these cells. Using this RNA, polynucleotide cDNA probes were prepared and hybridized to Clontech Atlas polynucleotide array filters as described previously. Hybridization of the cDNA probes to the respective immobilized DNAs can be visualized by autoradiography, and can be quantified using a phosphorescence imaging system. Tables 55-59 summarize the results of at least 2-3 independent experiments. It was found that treatment of RAW264.7 cells with LPS resulted in increased expression of over 60 polynucleotides encoding proteins including inflammatory proteins such as IL-1 β, Induced Nitric Oxide Synthase (iNOS), MIP-1 α, MIP-1 β, MIP-2 α, CD40 and various transcription factors. Comparison of the changes in polynucleotide expression induced by LPS, LTA and CpG DNA revealed that all three bacterial products increased the expression of pro-inflammatory polynucleotides to similar extents, such as iNOS, MIP-1 α, MIP-2 α, IL-1 β, IL-15, TNFR1 and NF- κ B (Table 57). Table 57 describes 19 polynucleotides that are up-regulated to a similar extent by bacterial products whose stimulation ratios do not differ more than 1.5-fold between the three bacterial products. Several polynucleotides are also down-regulated to a similar extent by LPS, LTA and CpG. It was also found that many polynucleotides were differentially regulated in response to these three bacterial products (Table 58), including many polynucleotides whose expression levels differed by more than 1.5-fold between one or more bacterial products. The expression of the polynucleotide was most affected by differences in LTA treatment compared to LPS or CpG, including overstimulation of Jun-D, Jun-B, Elk-1 and cyclins G2 and A1. Only some of the polynucleotides were more altered in expression by LPS or CpG treatment. LPS treatment was more able to increase the expression of several polynucleotides, including cAMP response element DNA binding protein (CRE-BP), interferon inducible protein 1, and CACCC frame binding protein BKLF, compared to LTA or CpG treatment. CpG treatment is more capable of increasing the expression of several polynucleotides, including Leukemia Inhibitory Factor (LIF) and protease ligase 1(PN-1), than LPS or LTA treatment. These results indicate that although the polynucleotides in response to LPS, LTA and CpGDNA stimulation of expression largely overlap, they also display different abilities to modulate certain polynucleotides.
[00151]Another polynucleotide array that may be used is the Human Operon array (the genome has an identification number of PRHU04-S1), which consists of approximately 14,000 spots of individual oligomers spotted in duplicate. Probes were prepared with 5. mu.g total RNA and labeled with Cy3 or Cy5 labeled dUTP. In these experiments, A549 epithelial cells were plated into 100mm tissue culture dishes at a density of 2.5X 10 per dish6Cells, incubated overnight, then incubated with 100ng/ml e.coli o 111: b4LPS was stimulated for 4 hours. Total RNA was isolated using RNAqueous (Ambion). DNA contamination was removed using a DNA removal kit (Ambion). Probes prepared from total RNA were purified and hybridized to printed glass slides, overnight at 42 ℃ and then washed. After washing, images were taken with a Perkin Elmer array scanner. The mean, median and background intensities of the spots were determined using image processing software (Imapolynucheotide 5.0, Marina Del Rey, Calif.). The background was removed using a "homemade" procedure. The program calculates the base intensity of each sub-cell as 10% and subtracts this value for each cell. Analysis was performed using polynuceotidization software (Redwood City, CA). Intermediate point intensities are obtained from a set of values for points within a slide and are to be measuredThis value was compared to the values for all slides in this experiment, thereby normalizing the intensity of each spot. The relative change between LPS treated cells and control cells can be seen in the table below. Table 60 describes a number of previously unreported changes that would be useful in diagnosing infection.
[00152] To confirm and assess the functional significance of these changes, the levels of selected mRNA and protein were estimated and quantified by densitometry. Northern blot hybridization using CD14, vimentin, and triple tetraprolin-specific probes demonstrated similar expression following stimulation with all three bacterial products (Table 60). The level of NO, an inflammatory mediator, was estimated using Griess's reagent and found to be comparable after 24 hours, and since the level of NO can be a marker for the enzymatic activity of the nitric oxide synthase iNOS, it was found that upregulation of iNOS expression was similar (table 59). Western blot analysis demonstrated that CpG preferentially stimulates leukemia inhibitory factor (LIF, a member of the cytokine IL-6 family) (Table 59). Additional confirmatory experiments demonstrated that LPS upregulation regulates TNF- α and IL-6 expression, as a result of ELISA assays; also up-regulated expression of MIP-2 alpha and IL-1 beta mRNA and down-regulated expression of DP-1 and cyclin D mRNA as a result of Northern blot hybridization. By testing the ability of the bacterial element to stimulate the production of proinflammatory cytokines in whole blood, the assay can be extended to in vitro systems that are more clinically relevant. Coli LPS, Salmonella typhimurium LPS and Staphylococcus aureus LTA were found to all stimulate similar production amounts of serum TNF- α and IL-1 β. CpG also stimulates the production of these cytokines, although at much lower levels, partially supporting the data for cell lines.
[00153] Table 55: a549 epithelial cells are differentiated by E coli O111: b4 an LPS upregulated polynucleotide. Studies with polynucleotide microarrays have shown that E coli O111: b4LPS (100ng/ml) increased the expression of many polynucleotides. LPS was incubated with a549 cells for 4 hours and RNA was isolated. cDNA probes labeled with Cy3/Cy5 were prepared from 5. mu.g total RNA and hybridized to a Human Operon array (PRHU 04). The intensity in unstimulated cells is shown in the third column of table 55. "ratio: LPS/control "column refers to the intensity of polynucleotide expression in LPS-stimulated cells divided by the intensity of non-stimulated cells.
| Registration number | Gene | Comparison: medium strength only | The ratio is: LPS/control |
| D87451 | Ring finger protein 10 | 715.8 | 183.7 |
| AF061261 | C3H type zinc finger protein | 565.9 | 36.7 |
| D17793 | Aldehyde-ketone reductase family 1, member C3 | 220.1 | 35.9 |
| M14630 | Thymosin alpha | 168.2 | 31.3 |
| AL049975 | Is unknown | 145.6 | 62.3 |
| L04510 | ADP-ribosylation factor domain protein 1, 64kD | 139.9 | 213.6 |
| U10991 | G2 protein | 101.7 | 170.3 |
| U39067 | Eukaryotic translation initiation factor 3, subunit 2 | 61.0 | 15.9 |
| X03342 | Ribosomal protein L32 | 52.6 | 10.5 |
| NM_004850 | Rho-associated, coiled-coil-containing protein kinase 2 | 48.1 | 11.8 |
| AK000942 | Is unknown | 46.9 | 8.4 |
| AB040057 | Serine/threonine protein kinase MASK | 42.1 | 44.3 |
| AB020719 | KIAA0912 protein | 41.8 | 9.4 |
| AB007856 | FEM-1-like death receptor binding proteins | 41.2 | 16.7 |
| J02783 | Procollagen-proline, 2-oxoglutarate 4-dioxygenase | 36.1 | 14.1 |
| AL137376 | Is unknown | 32.5 | 17.3 |
| AL137730 | Is unknown | 29.4 | 11.9 |
| D25328 | Phosphofructokinase, platelet | 27.3 | 8.5 |
| AF047470 | Malate dehydrogenase 2, NAD | 25.2 | 8.2 |
| M86752 | Stress-inducible phosphoprotein 1 | 22.9 | 5.9 |
| M90696 | Cathepsin S | 19.6 | 6.8 |
| AK001143 | Is unknown | 19.1 | 6.4 |
| AF038406 | NADH dehydrogenase | 17.7 | 71.5 |
| AK000315 | Hypothetical protein FLJ20308 | 17.3 | 17.4 |
| M54915 | Pim-1 oncogene | 16.0 | 11.4 |
| D29011 | Proteasome subunit, beta form, 5 | 15.3 | 41.1 |
| AK000237 | Membrane proteins of cholinergic synaptic vesicles | 15.1 | 9.4 |
| AL034348 | Is unknown | 15.1 | 15.8 |
| AL161991 | Is unknown | 14.2 | 8.1 |
| AL049250 | Is unknown | 12.7 | 5.6 |
| AL050361 | PTD017 protein | 12.6 | 13.0 |
| U74324 | RAB interactivity factor | 12.3 | 5.2 |
| M22538 | NADH dehydrogenase | 12.3 | 7.6 |
| D87076 | KIAA0239 protein | 11.6 | 6.5 |
| NM_006327 | Homologues of (yeast) endomitochondrial membrane ectopase 23 | 11.5 | 10.0 |
| AK001083 | Is unknown | 11.1 | 8.6 |
| AJ001403 | Mucin 5, subtype B, of the tracheobronchial type | 10.8 | 53.4 |
| M64788 | RAP1, GTPase activating protein 1 | 10.7 | 7.6 |
| X06614 | Retinoic acid receptor, alpha | 10.7 | 5.5 |
| U85611 | Calcium and integrin binding proteins | 10.3 | 8.1 |
| U23942 | Cytochrome P450, 51 | 10.1 | 10.2 |
| AL031983 | Is unknown | 9.7 | 302.8 |
| NM_007171 | Protein-mannosyltransferase | 9.5 | 6.5 |
| AK000403 | Hypothetical protein FLJ20396 | 9.5 | 66.6 |
| NM_002950 | Ribosomal receptor protein I | 9.3 | 35.7 |
| L05515 | cAMP response element binding protein CRE-BPa | 8.9 | 6.2 |
| X83368 | Phosphoinositide 3-kinases, catalytic, gamma polypeptides | 8.7 | 27.1 |
| M30269 | Nestin (enactin) | 8.7 | 5.5 |
| M91083 | Chromosome 11 open reading frame 13 | 8.2 | 6.6 |
| D29833 | Proline-rich proteins of saliva | 7.7 | 5.8 |
| AB024536 | Immunoglobulin superfamily containing leucine-rich repeats | 7.6 | 8.0 |
| U39400 | Chromosome 11 open reading frame 4 | 7.4 | 7.3 |
| AF028789 | unc119(c. elegans) homolog | 7.4 | 27.0 |
| NM_003144 | Signal sequence receptor, alpha (translocon-related protein alpha) | 7.3 | 5.9 |
| X52195 | Arachidonic acid 5-lipoxygenase activating protein | 7.3 | 13.1 |
| U43895 | Human growth factor-modulating tyrosine kinase substrates | 6.9 | 6.9 |
| L25876 | Cyclin-dependent kinase inhibitor 3 | 6.7 | 10.3 |
| L04490 | NADH dehydrogenase | 6.6 | 11.1 |
| Z18948 | S100 calcium binding protein | 6.3 | 11.0 |
| D10522 | Myristoylated alanine-rich protein kinase C substrates | 6.1 | 5.8 |
| NM_014442 | Sialic acid binding to Ig-like lectin 8 | 6.1 | 7.6 |
| U81375 | Solute carrier family 29 | 6.0 | 6.4 |
| AF041410 | Malignant tumor associated protein | 5.9 | 5.3 |
| U24077 | Killer cell immunoglobulin-like receptors | 5.8 | 14.4 |
| AL137614 | Hypothetical proteins | 4.8 | 6.8 |
| NM_002406 | Mannosyl (alpha-1, 3) -glycoprotein beta-1, 2-N-acetylglucosaminyltransferase | 4.7 | 5.3 |
| AB002348 | KIAA0350 protein | 4.7 | 7.6 |
| AF165217 | Promyoglobin conditional protein 4 (muscle) | 4.6 | 12.3 |
| Z14093 | Branched-chain ketoacid dehydrogenase E1, alpha Polypeptides | 4.6 | 5.4 |
| U82671 | Kallikrein | 3.8 | 44.5 |
| AL050136 | Is unknown | 3.6 | 5.0 |
| NM_005135 | Solute carrier family 12 | 3.6 | 5.0 |
| AK001961 | Hypothetical protein FLJ11099 | 3.6 | 5.9 |
| AL034410 | Is unknown | 3.2 | 21.3 |
| S74728 | antiquitin 1 | 3.1 | 9.2 |
| AL049714 | Ribosomal protein L34 pseudogene 2 | 3.0 | 19.5 |
| NM_014075 | PRO0593 protein | 2.9 | 11.5 |
| AF189279 | Phospholipase A2, class IIE | 2.8 | 37.8 |
| J03925 | Integrin,. alpha.M | 2.7 | 9.9 |
| NM_012177 | F-box protein Fbx5 | 2.6 | 26.2 |
| NM_004519 | Voltage-gated potassium channel, KQT-like subfamily, member 3 | 2.6 | 21.1 |
| M28825 | CD1A antigen, polypeptide | 2.6 | 16.8 |
| X16940 | Actin, gamma 2, smooth intestinal muscle | 2.4 | 11.8 |
| X03066 | Class II major histocompatibility complex, DO beta | 2.2 | 36.5 |
| AK001237 | Hypothetical protein FLJ10375 | 2.1 | 18.4 |
| AB028971 | KIAA1048 protein | 2.0 | 9.4 |
| AL137665 | Is unknown | 2.0 | 7.3 |
[00154] Table 56: a549 epithelial cells are differentiated by E coli O111: b4 a polynucleotide downregulated by LPS. Studies with polynucleotide microarrays have shown that E coli O111: b4LPS (100ng/ml) reduced the expression of many polynucleotides in A549 cells. LPS was incubated with a549 cells for 4 hours and RNA was isolated. cDNA probes labeled with Cy3/Cy5 were prepared from 5. mu.g total RNA and hybridized to a Human Operon array (PRHU 04). The intensity in the unstimulated cells is shown in the third column of Table 5. "ratio: LPS/control "column refers to the intensity of polynucleotide expression in LPS-stimulated cells divided by the intensity of non-stimulated cells.
| Registration number | Gene | Comparison: medium strength only | The ratio is: LPS/control |
| NM_017433 | Myosin IIIA | 167.8 | 0.03 |
| X60484 | H4 Histone family member E | 36.2 | 0.04 |
| X60483 | H4 Histone family member D | 36.9 | 0.05 |
| AF151079 | Hypothetical proteins | 602.8 | 0.05 |
| M96843 | DNA-binding arrestin 2, dominant negative helix-loop-helix protein | 30.7 | 0.05 |
| S79854 | Deiodinase, iodothyronine, type III | 39.4 | 0.06 |
| AB018266 | matrin 3 | 15.7 | 0.08 |
| M33374 | NADH dehydrogenase | 107.8 | 0.09 |
| AF005220 | mRNA, partial cds of human NUP98-HOXD13 fusion protein | 105.2 | 0.09 |
| Z80783 | H2B Histone family, member L | 20.5 | 0.10 |
| Z46261 | H3 Histone family, member A | 9.7 | 0.12 |
| Z80780 | H2B Histone family, member H | 35.3 | 0.12 |
| U33931 | Erythrocyte membrane protein band 7.2(stomatin) | 18.9 | 0.13 |
| M60750 | H2B Histone family, member A | 35.8 | 0.14 |
| Z83738 | H2B Histone family, member E | 19.3 | 0.15 |
| Y14690 | Collagen, type V, alpha 2 | 7.5 | 0.15 |
| M30938 | XRCC5, X-ray repair, supplementation of defective repair function of Chinese hamster cells | 11.3 | 0.16 |
| L36055 | Eukaryotic translation initiation factor 4E binding proteins | 182.5 | 0.16 |
| Z80779 | H2B Histone family, member G | 54.3 | 0.16 |
| AF226869 | 5(3) -deoxyribonuclease; RB-related KRAB repressors | 7.1 | 0.18 |
| D50924 | KIAA0134 gene product | 91.0 | 0.18 |
| AL133415 | Vimentin | 78.1 | 0.19 |
| AL050179 | Tropomyosin 1 (. alpha.) | 41.6 | 0.19 |
| AJ005579 | RD element | 5.4 | 0.19 |
| M80899 | AHNAK nuclear protein | 11.6 | 0.19 |
| NM_004873 | BCL2 relevance athanogene 5 | 6.2 | 0.19 |
| X57138 | H2A Histone family, member N | 58.3 | 0.20 |
| AF081281 | Lysophospholipase I | 7.2 | 0.22 |
| U96759 | von Hippel-Linau binding protein I | 6.6 | 0.22 |
| U85977 | Human ribosomal protein L12 pseudogene, part of cds | 342.6 | 0.22 |
| D13315 | Glyoxalases | 7.5 | 0.22 |
| AC003007 | Is unknown | 218.2 | 0.22 |
| AB032980 | RU2S | 246.6 | 0.22 |
| U40282 | Integrin-associated kinases | 10.1 | 0.22 |
| U81984 | Endothelial PAS domain protein 1 | 4.7 | 0.23 |
| X91788 | Chloride channels, nucleotide sensitive, 1A | 9.6 | 0.23 |
| AF018081 | Collagen, type XVIII, alpha 1 | 6.9 | 0.24 |
| L31881 | Nuclear factor I/X (CCAAT-binding transcription factor) | 13.6 | 0.24 |
| X61123 | B cell translocation Gene 1, antiproliferation | 5.3 | 0.24 |
| L32976 | Mitogen-activated protein kinase 11 | 6.3 | 0.24 |
| M27749 | Immunoglobulin lambda-like polypeptide 3 | 5.5 | 0.24 |
| X57128 | H3 Histone family, member C | 9.0 | 0.25 |
| X80907 | Phosphoinositide-3-kinases, regulatory subunits, polypeptide 2 | 5.8 | 0.25 |
| Z34282 | Human mucin (MAR11) MUC5AC mRNA (in part) | 100.6 | 0.26 |
| X00089 | H2A grouped eggsWhite family, member M | 4.7 | 0.26 |
| AL035252 | CD 39-like 2 | 4.6 | 0.26 |
| X95289 | PERB11 family members of the MHC class I region | 27.5 | 0.26 |
| AJ001340 | U3 snoRNP-related 55kDa protein | 4.0 | 0.26 |
| NM_014161 | HSPC071 protein | 10.6 | 0.27 |
| U60873 | Is unknown | 6.4 | 0.27 |
| X91247 | Thioredoxin reductase 1 | 84.4 | 0.27 |
| AK001284 | Hypothetical protein FLJ10422 | 4.2 | 0.27 |
| U90840 | Synovial sarcoma, X breakpoint 3 | 6.6 | 0.27 |
| X53777 | Ribosomal protein L17 | 39.9 | 0.27 |
| AL035067 | Is unknown | 10.0 | 0.28 |
| AL117665 | DKFZP586M1824 protein | 3.9 | 0.28 |
| L14561 | ATP enzyme, calcium ion transport, plasma Membrane 1 | 5.3 | 0.28 |
| L19779 | H2A Histone family, member O | 30.6 | 0.28 |
| AL049782 | Is unknown | 285.3 | 0.28 |
| X00734 | Tubulin, beta, 5 | 39.7 | 0.29 |
| AK001761 | Retinoic acid inducibility 3 | 23.7 | 0.29 |
| U72661 | ninjurin 1 | 4.4 | 0.29 |
| S48220 | Deiodinase, iodothyronine, type I | 1,296.1 | 0.29 |
| AF025304 | EphB2 | 4.5 | 0.30 |
| S82189 | Chymotrypsin C | 4.1 | 0.30 |
| Z80782 | H2B Histone family, member K | 31.9 | 0.30 |
| X68194 | Synaptic vesicle protein-like proteins | 7.9 | 0.30 |
| AB028869 | Is unknown | 4.2 | 0.30 |
| AK000761 | Is unknown | 4.3 | 0.30 |
[00155] Table 57: polynucleotides expressed to a similar extent after stimulation with the bacterial products LPS, LTA and CpG DNA. Bacterial products (100ng/ml Salmonella typhimurium LPS, 1. mu.g/ml Staphylococcus aureus LTA or 1. mu.M CpG) were found to efficiently induce the expression of several polynucleotides. The bacterial products were incubated with RAW cells for 4 hours, RNA was isolated, converted to labeled cDNA probes, and hybridized to an Atlas array. The intensity in control cells that were not stimulated is shown in the second column. "ratio: LPS/LTA/CpG: control "column" refers to the result of dividing the intensity of polynucleotide expression in cells stimulated with bacterial product by the intensity of unstimulated cells.
| Registration number | Control non-stimulated intensity | Ratio LPS: control | The ratio LTA: control | Ratio CpG: control | Protein/polynucleotide |
| M15131 | 20 | 82 | 80 | 55 | IL-1β |
| M57422 | 20 | 77 | 64 | 90 | Triple tetra proline basic protein |
| X53798 | 20 | 73 | 77 | 78 | MIP-2α |
| M35590 | 188 | 50 | 48 | 58 | MIP-1β |
| L28095 | 20 | 49 | 57 | 50 | ICE |
| M87039 | 20 | 37 | 38 | 45 | iNOS |
| X57413 | 20 | 34 | 40 | 28 | TGFβ |
| X15842 | 20 | 20 | 21 | 15 | c-rel proto-cancer polynucleotides |
| X12531 | 489 | 19 | 20 | 26 | MIP-1α |
| U14332 | 20 | 14 | 15 | 12 | IL-15 |
| M59378 | 580 | 10 | 13 | 11 | TNFR1 |
| U37522 | 151 | 6 | 6 | 6 | TRAIL |
| M57999 | 172 | 3.8 | 3.5 | 3.4 | NF-κB |
| U36277 | 402 | 3.2 | 3.5 | 2.7 | I-kappa B (alpha subunit) |
| X76850 | 194 | 3 | 3.8 | 2.5 | MAPKAP-2 |
| U06924 | 858 | 2.4 | 3 | 3.2 | Stat1 |
| X14951 | 592 | 2 | 2 | 2 | CD18 |
| X60671 | 543 | 1.9 | 2.4 | 2.8 | NF-2 |
| M34510 | 5970 | 1.6 | 2 | 1.4 | CD14 |
| X51438 | 2702 | 1.3 | 2.2 | 2.0 | Vimentin |
| X68932 | 4455 | 0.5 | 0.7 | 0.5 | c-Fms |
| Z21848 | 352 | 0.5 | 0.6 | 0.6 | DNA polymerase |
| X70472 | 614 | 0.4 | 0.6 | 0.5 | B-myb |
[00156] Table 58: a polynucleotide differentially regulated by the bacterial products LPS, LTA and CpG DNA. Bacterial products (100ng/ml Salmonella typhimurium LPS, 1. mu.g/ml Staphylococcus aureus LTA or 1. mu.M CpG) were found to efficiently induce the expression of several polynucleotides. The bacterial products were incubated with RAW cells for 4 hours, RNA was isolated, converted to labeled cDNA probes, and hybridized to an Atlas array. The intensity in control cells that were not stimulated is shown in the second column. "ratio: LPS/LTA/CpG: control "column" refers to the result of dividing the intensity of polynucleotide expression in cells stimulated with bacterial product by the intensity of unstimulated cells.
| Registration number | Control non-stimulated intensity | Ratio LPS: control | The ratio LTA: control | Ratio CpG: control | Protein/polynucleotide |
| X72307 | 20 | 1.0 | 23 | 1.0 | Hepatocyte growth factor |
| L38847 | 20 | 1.0 | 21 | 1.0 | Transmembrane kinase ligand for liver cancer |
| L34169 | 393 | 0.3 | 3 | 0.5 | Thrombopoietin |
| J04113 | 289 | 1 | 4 | 3 | Nur77 |
| Z50013 | 20 | 7 | 21 | 5 | H-ras proto-carcinoma polynucleotides |
| X84311 | 20 | 4 | 12 | 2 | Cyclin A1 |
| U95826 | 20 | 5 | 14 | 2 | Cyclin G2 |
| X87257 | 123 | 2 | 4 | 1 | Elk-1 |
| J05205 | 20 | 18 | 39 | 20 | Jun-D |
| J03236 | 20 | 11 | 19 | 14 | Jun-B |
| M83649 | 20 | 71 | 80 | 42 | Fas 1 receptor |
| M83312 | 20 | 69 | 91 | 57 | CD40L receptor |
| X52264 | 20 | 17 | 23 | 9 | ICAM-1 |
| M13945 | 573 | 2 | 3 | 2 | Pim-1 |
| U60530 | 193 | 2 | 3 | 3 | Mad-related proteins |
| D10329 | 570 | 2 | 3 | 2 | CD7 |
| X06381 | 20 | 55 | 59 | 102 | Leukemia Inhibitory Factor (LIF) |
| X70296 | 20 | 6.9 | 13 | 22 | Protease connexin 1(PN-1) |
| U36340 | 20 | 38 | 7 | 7 | CACCC frame binding protein BKLF |
| S76657 | 20 | 11 | 6 | 7 | CRE-BPI |
| U19119 | 272 | 10 | 4 | 4 | Interferon inducible protein 1 |
[00157] Table 59: the array data of tables 57 and 58 were confirmed. a) Total RNA was isolated from unstimulated RAW macrophages and cells treated with 100ng/ml Salmonella typhimurium LPS, 1. mu.g/ml Staphylococcus aureus LTA, 1. mu.M CpG DNA or medium alone for 4 hours, subjected to Northern blot hybridization, probed for GAPDH, CD14, vimentin and triply tetraprolin on the membrane as described previously [ Scott et al ]. The hybridization intensity of Northern blots was compared with GAPDH to find inconsistencies in loading. These experiments were repeated at least three times, and the data presented were the mean levels of the various conditions (measured using densitometry) versus the medium ± standard deviation.
b) RAW264.7 cells were stimulated with 100ng/ml Salmonella typhimurium LPS, 1. mu.g/ml Staphylococcus aureus LTA, 1. mu.MCPG DNA or medium alone for 24 hours. Protein lysates were prepared, separated on SDS-PAGE gels, and Western blot hybridization was performed to detect LIF (R & D system). These experiments were repeated at least three times, and the data presented were the level of LIF relative to the medium (measured using densitometry) ± standard deviation.
c) RAW macrophages were stimulated with 100ng/ml Salmonella typhimurium LPS, 1. mu.g/ml Staphylococcus aureus LTA, 1. mu.M pG DNA or medium alone for 24 hours, cell supernatants were collected and the amount of NO formed in the supernatants was measured using Griess reagent, estimated using the accumulation of nitrite, a stable NO metabolite, as described previously [ Scott et al ]. Data presented are mean ± standard deviation of three experiments.
| Product of | Relative horizontal | |||
| Untreated | LPS | LTA | CpG | |
| CD14a | 1.0 | 2.2±0.4 | 1.8±0.2 | 1.5±0.3 |
| Vimentina | 1.0 | 1.2±0.07 | 1.5±0.05 | 1.3±0.07 |
| Triple tetra proline basic proteina | 1.0 | 5.5±0.5 | 5.5±1.5 | 9.5±1.5 |
| LIFb | 1.0 | 2.8±1.2 | 2.7±0.6 | 5.1±1.6 |
| NOc | 8±1.5 | 47±2.5 | 20±3 | 21±1.5 |
[00158] Table 60: a549 gene expression pattern in human epithelial cells upregulated by bacterial signaling molecules (LPS). Studies with polynucleotide microarrays have shown that E coli O111: b4LPS (100ng/ml) increased the expression of many polynucleotides in A549 cells. LPS was incubated with a549 cells for 4 hours and RNA was isolated. Cy3/Cy5 labeled cDNA probes were prepared with 5. mu.g total RNA and hybridized to a Human Operon array (PRHU 04). The intensity in unstimulated cells is shown in the third column of table 55. These examples of changes in polynucleotide expression in cells stimulated with LPS represent greater than 2-fold changes in the intensity levels compared to untreated cells.
| Registration number | Gene |
| AL050337 | Interferon gamma receptor 1 |
| U05875 | Interferon gamma receptor 2 |
| NM_002310 | Leukemia inhibitory factor receptors |
| U92971 | Coagulation factor II (thrombin) receptor-like 2 |
| Z29575 | Tumor necrosis factor receptor superfamily member 17 |
| L31584 | Chemokine receptor 7 |
| J03925 | cAMP response element binding proteins |
| M64788 | RAP1, GTPase activating protein |
| NM_004850 | Rho-associated kinase 2 |
| D87451 | Ring finger protein 10 |
| AL049975 | Is unknown |
| U39067 | Eukaryotic translation initiation factor 3, subunit 2 |
| AK000942 | Is unknown |
| AB040057 | Serine/threonine protein kinase MASK |
| AB020719 | KIAA0912 protein |
| AB007856 | FEM-1-like death receptor binding proteins |
| AL137376 | Is unknown |
| AL137730 | Is unknown |
| M90696 | Cathepsin S |
| AK001143 | Is unknown |
| AF038406 | NADH dehydrogenase |
| AK000315 | Hypothetical protein FLJ20308 |
| M54915 | Pim-1 oncogene |
| D29011 | Proteasome subunit, beta form, 5 |
| AL034348 | Is unknown |
| D87076 | KIAA0239 protein |
| AJ001403 | Tracheal bronchial mucin 5, subtype B, |
| J03925 | integrin,. alpha.M |
Example 10
Altering signaling to protect against bacterial infection
[00159]Salmonella typhimurium strain SL1344 was obtained from the American type culture Collection (ATCC; Manassas, Va.) and cultured in Luria-Bertani (LB) liquid medium. To effect macrophage infection, a frozen glycerol stock was inoculated into 10ml LB in a 125ml shake flask and cultured overnight at 37 ℃ with shaking to a stable phase. RAW264.7 cells (1X 10)5Individual cells/well) were seeded into 24-well plates. The bacteria were diluted with medium until a nominal combined infection (MOI) of approximately 100 was obtained, centrifuged onto a monolayer of cells by centrifugation at 1000rpm for 10 minutes to synchronize infection, and then allowed to infect at 37 ℃ in 5% CO2The incubator was continued for 20 minutes. Cells were washed 3 times with PBS to remove extracellular bacteria, and then incubated in DMEM + 10% FBS containing 100. mu.g/ml gentamicin (Sigma, St. Louis, Mo.) to killAny residual extracellular bacteria, avoiding reinfection. After two hours, the gentamicin concentration was reduced to 10 μ g/ml and maintained at this value throughout the analysis. Cells were pretreated for 30 min with inhibitors with the following concentrations before infection: 50 μ MPD98059(Calbiochem), 50 μ M U0126 (Promega), 2mM Diiodo (DPI), 250 μ M acetovanidone (apocynin, Aldrich), 1mM ascorbic acid (Sigma), 30mM N-acetylcysteine (Sigma) and 2mM NGL-monomethyl arginine (L-NMMA, Molecular Probe) or 2mM NGD-monomethyl arginine (D-NMMA, Molecular Probe). Fresh inhibitor was added immediately, 2 hours and 6-8 hours after infection to ensure efficacy. Control cells were treated with an equal volume of dimethyl sulfoxide (DMSO) per ml of medium. The intracellular survival/replication of salmonella typhimurium SL1344 was determined using the gentamicin resistance assay, as described previously. Briefly, at 2 and 24 hours post-infection, cells were washed twice with PBS to remove gentamicin, lysed with PBS containing 1% TritonX-100/0.1% SDS, and the number of bacteria inside the cells was calculated from the number of colonies on LB agar plates. Under these infection conditions, macrophages contain an average of one bacterium per cell as estimated by standard plate counts, and thus can be analyzed 24 hours after infection. Bacterial fibrillation (filamentation) is associated with bacterial stress (stress). NADPH oxidase and iNOS can be activated by MEK/ERK signaling. The results (Table 61) clearly demonstrate that altering cell signaling is one way by which Salmonella intracellularis infection can be addressed. Since bacteria upregulate multiple genes in human cells, this strategy of blocking signal transduction represents a common approach to anti-infective therapy.
[00160] Table 61: effect of the signaling molecule MEK on intracellular bacteria in IFN- γ sensitized RAW cells.
| Treatment ofa | Effectb |
| 0 | Is free of |
| MEK inhibitor U0126 | Reducing bacterial fibrillation (bacterial stress)cIncreasing the number of Salmonella typhimurium in cells |
| MEK inhibitor PD98059 | Reducing bacterial fibrillation (bacterial stress)cIncreasing the number of Salmonella typhimurium in cells |
| NADPH oxidase inhibitorsd | Reducing bacterial fibrillation (bacterial stress)cIncreasing the number of Salmonella typhimurium in cells |
Example 11
Antiviral activity
[00161]SDF-1 is a C-X-C chemokine, which is a natural ligand of the HIV-1 co-receptor CXCR-4. In order to inhibit the replication of HIV-1, the chemokine receptors CXCR-4 and CCR5 are considered as possible targets. The crystal structure of SDF-1 exhibits antiparallel beta-sheets and a positively charged surface, characteristics that are important for its binding to the negatively charged extracellular loops of CXCR-4. These findings suggest that chemokine derivatives, small CXCR4 antagonists, or agonists that mimic the structural or ionic properties of chemokines may be useful agents for the treatment of X4 HIV-1 infection. The cationic peptides were found to inhibit SDF-1 induced T cell migration, suggesting that these peptides may act as CXCR-4 antagonists. Migration analysis was performed as follows. Human Jurkat T cells were resuspended to 5X 10 using chemotactic medium (RPMI 1640/10mM Hepes/0.5% BSA)6Individual cells/ml. Migration analysis was performed in 24-well plates using a5 μm polycarbonate Transwell insert (Costar). Briefly, peptides or controls were diluted with chemotactic medium and placed in the lower chamber while 0.1ml of cells (5X 10)6Pieces/ml) was added to the upper chamber. After 3 hours at 37 ℃, the number of cells migrating into the lower chamber was determined by flow cytometry. The culture in the lower chamber took 30 seconds to traverse the FACscan, opening the front and side diffusers to expel cell debris. The number of viable cells was compared with the "100% migration control", for which 5X 105Individual cells were pipetted directly into the lower chamber and then counted for 30 seconds using a FACscan. The results indicate that the addition of the peptide results in the inhibition of migration of human Jurkat T cells (table 62), likely because the expression of CXCR-4 was affected (tables 63 and 64).
[00162] Table 62: the peptides inhibit migration of human Jurkat T cells:
| migration (%) | ||||
| Experiment of | Positive control | SDF-1(100ng/ml) | SDF-1+SEQ ID 1(50μg/ml) | Negative control |
| 1 | 100% | 32% | 0% | <0.01% |
| 2 | 100% | 40% | 0% | 0% |
[00163] Table 63: polynucleotide array data corresponding to table 56:
| polynucleotides/proteins | Polynucleotide function | Intensity of non-stimulation | Ratio peptide: is not stimulated | Registration number |
| CXCR-4 | Chemokine receptors | 36 | 4 | D87747 |
[00164] Table 64: FACs data corresponding to tables 62 and 63:
| peptides | Concentration (μ g/ml) | Fold increase in protein expression CXCR-4 |
| SEQ ID NO:1 | 10 | Has no change |
| SEQ ID NO:1 | 50 | 1.3±0.03 |
| SEQ ID NO:1 | 100 | 1.6±0.23 |
| SEQ ID NO:3 | 100 | 1.5±0.2 |
[00164] While the invention has been described by reference to a preferred embodiment, it should be understood that various changes may be made without departing from the spirit of the invention. Accordingly, the invention is not limited except as by the appended claims.
Claims (20)
1. Comprising the general formula X1X2X3IX4PX4IPX5X2X1(SEQ ID NO: 4) wherein X1Is one or two of R, L or K, X2Is C, S or A, X3Is an R or P, X4Is an A or V, X5Is a V or W.
2. The cationic peptide according to claim 1, wherein the peptide is selected from the group consisting of: LLCRIVPVIPWCK (SEQ ID NO: 5), LRCPIAPVIPVCKK (SEQ ID NO: 6), KSRIVPAIPVSLL (SEQ ID NO: 7), KKSPIAPAIPWSR (SEQ ID NO: 8), RRARIVPAIPVARR (SEQ ID NO: 9) and LSRIAPAIPWAKL (SEQ ID NO: 10).
3. The cationic peptide according to claim 1, wherein the peptide has anti-inflammatory activity.
4. The cationic peptide according to claim 1, wherein the peptide has anti-sepsis activity.
5. The cationic peptide according to any of claims 1 to 4, wherein said peptide comprises at least one amino acid of the D-enantiomer.
6. The cationic peptide according to any of claims 1-4, wherein said peptide is cyclic.
7. The cationic peptide according to any of claims 1-4, wherein said peptide is inverted.
8. The cationic peptide according to claim 5, wherein said peptide is cyclic.
9. The cationic peptide according to claim 5, wherein said peptide is inverted.
10. The cationic peptide according to claim 6, wherein said peptide is inverted.
11. The cationic peptide according to claim 8, wherein said peptide is inverted.
12. Consisting of SEQ ID NO: 4 in the manufacture of a medicament for stimulating innate immunity in an individual, wherein a therapeutically effective amount of the peptide is comprised in the medicament.
13. Use according to claim 12, wherein said innate immunity is evidenced by activation of the MAP kinase pathway.
14. Use according to claim 13, wherein the MAP kinase is MEK and/or ERK.
15. The use according to claim 12, wherein said peptide has anti-inflammatory activity.
16. Use according to claim 12, wherein the peptide has anti-sepsis activity.
17. Use according to claim 12, wherein said peptide comprises at least one amino acid of the D-enantiomer.
18. Use according to claim 12, wherein said peptide is cyclic.
19. The cationic peptide according to claim 1 or 2, wherein the amino acids of the peptide are in the D configuration and are linked in a reverse sequence.
20. The cationic peptide according to claim 19, wherein all amino acids of the peptide are the D enantiomer.
Applications Claiming Priority (3)
| Application Number | Priority Date | Filing Date | Title |
|---|---|---|---|
| US33663201P | 2001-12-03 | 2001-12-03 | |
| US60/336,632 | 2001-12-03 | ||
| PCT/CA2002/001830 WO2003048383A2 (en) | 2001-12-03 | 2002-12-02 | Effectors of innate immunity |
Publications (2)
| Publication Number | Publication Date |
|---|---|
| HK1075677A1 HK1075677A1 (en) | 2005-12-23 |
| HK1075677B true HK1075677B (en) | 2008-08-22 |
Family
ID=
Similar Documents
| Publication | Publication Date | Title |
|---|---|---|
| CN100357324C (en) | Effectors of innate immunity | |
| US7507787B2 (en) | Effectors of innate immunity | |
| García et al. | Identification of a novel, multifunctional β-defensin (human β-defensin 3) with specific antimicrobial activity: its interaction with plasma membranes of Xenopus oocytes and the induction of macrophage chemoattraction | |
| EP1410018A1 (en) | Method for detecting modultators of notch signalling | |
| Moran et al. | Towards a transcriptome definition of microglial cells | |
| US20070134261A1 (en) | Effectors of innate immunity | |
| EP1991251A1 (en) | Use of tlr3 agonists for the treatment of neurodegenerative disorders | |
| US20050201975A1 (en) | Medical treatment | |
| US7687454B2 (en) | Effectors of innate immunity determination | |
| US20070190533A1 (en) | Effectors of innate immunity | |
| HK1075677B (en) | Effectors of innate immunity | |
| KR20070033314A (en) | Methods of Stimulating Innate Immunity Using Cationic Peptides | |
| HK1121196A (en) | Effectors of innate immunity | |
| AU2007201885A1 (en) | Effectors of innate immunity | |
| HK1098969A (en) | Methods of stimulating innate immunity using cationic peptides | |
| WO2004087195A2 (en) | Therapeutic use of modulators of notch and/or kruppel-like factors | |
| US20030166599A1 (en) | Uses of tgap7 for the modulation of leucocyte activation | |
| JPWO2003070947A1 (en) | Apoptosis-promoting substance and inhibitory substance interacting with CGI-94 and screening method thereof | |
| AU2002355670A1 (en) | Modulators of notch signalling for use in immunotherapy |